Language selection

Search

Patent 3071817 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3071817
(54) English Title: METHODS AND COMPOSITIONS FOR TREATMENT OF AMYLOID DEPOSITION DISEASES
(54) French Title: METHODES ET COMPOSITIONS POUR LE TRAITEMENT DES MALADIES A DEPOTS AMYLOIDES
Status: Allowed
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 39/395 (2006.01)
  • A61P 43/00 (2006.01)
(72) Inventors :
  • LENTZSCH, SUZANNE (United States of America)
(73) Owners :
  • THE TRUSTEES OF COLUMBIA UNIVERSITY IN THE CITY OF NEW YORK (United States of America)
(71) Applicants :
  • THE TRUSTEES OF COLUMBIA UNIVERSITY IN THE CITY OF NEW YORK (United States of America)
(74) Agent: MBM INTELLECTUAL PROPERTY AGENCY
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2018-07-24
(87) Open to Public Inspection: 2019-02-07
Examination requested: 2020-01-31
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2018/043374
(87) International Publication Number: WO2019/027721
(85) National Entry: 2020-01-31

(30) Application Priority Data:
Application No. Country/Territory Date
62/539,821 United States of America 2017-08-01
62/637,609 United States of America 2018-03-02

Abstracts

English Abstract


Methods and pharmaceutical compositions for treatment of amyloid deposition
diseases using chimeric (e.g., mouse-
human) antibody are disclosed, including a method for treating amyloid
deposition diseases with cardiac involvement by administering
pharmaceutical compositions comprising a chimeric anti-amyloid fibril
antibody. The methods herein can improve myocardial function
in patients diagnosed with light chain amyloid light chain amyloidosis (ALA)
having a cardiac involvement in as little as three weeks
after treatment.


French Abstract

L'invention concerne des méthodes et des compositions pharmaceutiques pour le traitement des maladies à dépôts amyloïdes utilisant un anticorps chimérique (p. ex., murin-humain), dont une méthode de traitement de maladies à dépôts amyloïdes ayant une composante cardiaque par administration de compositions pharmaceutiques comprenant un anticorps chimérique anti-fibrilles amyloïdes. Les méthodes selon l'invention peuvent améliorer la fonction myocardique dès trois semaines après le traitement.chez les patients chez lesquels on a diagnostiqué une amylose à chaînes légères (AL) caractérisée par des dépôts amyloïdes à chaînes légères.

Claims

Note: Claims are shown in the official language in which they were submitted.


CLAIMS
What is claimed is:
1. A method of treating an amyloid deposition disease in a human patient
in need of such treatment which comprises administering to the patient
a pharmaceutical composition comprising a chimeric mouse-human
antibody which comprises a V K region comprising SEQ ID NO: 47 and a
V H region comprising SEQ ID NO: 48 in a dose effective to treat said
amyloid deposition disease, together with a pharmaceutically
acceptable carrier.
2. The method of claim 1, wherein the amyloid deposition disease is
primary amyloidosis.
3. The method of claim 2, wherein the chimeric antibody is administered in
a dose of about 500 mg/m2 or less.
4. The method of claim 2, wherein the effective dose of a chimeric
antibody is about 2,200 mg.
5. The method of claim 2, wherein the effective dose of a chimeric
antibody is about 1-50 mg/kg.
6. The method of claim 2, wherein the primary amyloidosis comprises
involvement of at least one organ or tissue selected from the group
consisting of heart, kidneys, liver, lung, gastrointestinal tract, nervous
system, muscular skeletal system, soft tissue, and skin.
7. The method of claim 2, wherein the dose is effective to cause signs of a

therapeutic response in less than 5 weeks.
8. The method of claim 2, wherein the patient shows signs of a
therapeutic response within about a week or less.
9. The method of claim 6, wherein the primary amyloidosis comprises
involvement of the heart.
76

10. The method of claim 7, wherein the dose is effective to cause the
patient's N-terminal pro b-type natriuretic peptide (NT-proBNP) level to
decrease by at least about 30% compared to baseline levels following
administration of the chimeric antibody.
11. The method of claim 7, wherein the dose is effective to cause the
patient's N-terminal pro b-type natriuretic peptide (NT-proBNP) level to
decrease by at least about 40% compared to baseline levels following
administration of the chimeric antibody.
12. The method of claim 7, wherein the dose is effective to cause the
patient's N-terminal pro b-type natriuretic peptide (NT-proBNP) level to
be less than about 9100 ng/L following administration of the antibody.
13. The method of claim 7, wherein the dose is effective to cause the
patient's N-terminal pro b-type natriuretic peptide (NT-proBNP) level to
be less than about 8000 ng/L following administration of the chimeric
antibody.
14. The method of claim 7, wherein the dose is effective to cause the
patient's N-terminal pro b-type natriuretic peptide (NT-proBNP) level to
be less than about 7000 ng/L following administration of the chimeric
antibody.
15. The method of claim 7, wherein the dose is effective to cause the
patient's N-terminal pro b-type natriuretic peptide (NT-proBNP) level to
be less than about 6000 ng/L following administration of the chimeric
antibody.
16. The method of claim 7, wherein the dose is effective to cause the
patient's N-terminal pro b-type natriuretic peptide (NT-proBNP) level to
be less than about 5000 ng/L following administration of the chimeric
antibody.
17. The method of claim 7, wherein the patient was classified as New York
Heart Association (NYHA) Functional Classification class 11 or 111 prior to
77

administration of the chimeric antibody and is classified as class I
following administration of the chimeric antibody.
18. The method of claim 6, wherein the primary amyloidosis comprises
involvement of the kidneys.
19. The method of claim 18, wherein the dose is effective to cause the
patient's urine protein level to decrease by at least about 30%
compared to baseline levels following administration of the antibody.
20. The method of claim 18, wherein the dose is effective to cause the
patient's urine protein level to decrease by at least about 40%
compared to baseline levels following administration of the antibody.
21. The method of claim 18, wherein the dose is effective to cause the
patient's urine protein to be less than about 7000 mg/24 hours following
administration of the antibody.
22. The method of claim 18, wherein the dose is effective to cause the
patient's urine protein to be less than about 6000 mg/24 hours following
administration of the antibody.
23. The method of claim 18, wherein the dose is effective to cause the
patient's urine protein to be less than about 5000 mg/24 hours following
administration of the antibody.
24. The method of claim 18, wherein the dose is effective to cause the
patient's urine protein is less than about 4000 mg/24 hours following
administration of the antibody.
25. The method of claim 1, wherein administration of the antibody does not
cause any serious adverse events.
26. A method of detecting the presence of amyloid deposits in a patient
suspected of having such deposits which comprises administering to
the patient an amount of a chimeric mouse-human antibody which
comprises a V K region comprising SEQ ID NO: 47 and a V H region
comprising SEQ ID NO: 48 and having a detectable label attached
78

thereto, the amount of antibody administered being sufficient to allow
detection of amyloid deposits if such are present.
27. The method of claim 25, wherein the detectable label is 124I.
28. A method of treating a human patient with an amyloid deposit disease
comprising administering to said patient a therapeutically effective
amount of a chimeric 11-1F4 antibody less frequently than once per
month.
29. The method of claim 27, wherein the chimeric 11-1F4 antibody
comprises a constant region is derived from a human IgG1.
30. The method of claim 27, wherein the amyloid deposition disease is
primary light chain (AL) amyloidosis.
31. The method of claim 29, wherein the AL amyloidosis is comprises
aggregates of lambda light chain fibrils.
32. The method of claim 30, wherein the presence of aggregates of lambda
light chain fibrils is significantly reduced following administration of the
chimeric 11-1F4 antibody.
33. The method of claim 27, wherein the effective amount of the chimeric
11-1F4 antibody is 500 mg/m2 or less.
34. The method of claim 27, wherein the effective amount of the chimeric
11-1F4 antibody is about 2,200 mg.
35. The method of claim 27, wherein the effective amount of the chimeric
11-1F4 antibody is about 1-50 mg/kg.
36. The method of claim 27, wherein the amount is effective to cause the
patient to show signs of a therapeutic response in less than 5 weeks.
37. The method of claim 27, wherein the amount is effective to cause the
patient to show signs of a therapeutic response within about a week or
less.
79

38. The method of claim 27, wherein the therapeutically effective amount of

the chimeric 11-1F4 antibody is administered once every two months.
39. The method of claim 27, wherein the therapeutically effective amount of

the chimeric 11-1F4 antibody is administered once every three months.
40. The method of claim 27, wherein the therapeutically effective amount of

the chimeric 11-1F4 antibody is administered once every four months.
41. The method of claim 27, wherein the therapeutically effective amount of

the chimeric 11-1F4 antibody is administered once every five months.
42. The method of claim 27, wherein the therapeutically effective amount of

the chimeric 11-1F4 antibody is administered once every six months.
43. The method of claim 27, wherein the therapeutically effective amount of

the chimeric 11-1F4 antibody is administered biannually.
44. A method of treating a patient with primary light chain (AL)
amyloidosis
involving the heart comprising administering to said patient a dose of a
chimeric 11-1F4 antibody sufficient to cause at least a 30% reduction in
N-terminal pro b-type natriuretic peptide (NT-proBNP) level for said
patient following administration of the chimeric 11-1F4 antibody
compared to pre-treatment level.
45. The method of claim 43, wherein the AL amyloidosis is refractory.
46. The method of claim 43, wherein the chimeric 11-1F4 antibody
comprises a constant region is derived from a human IgG1.
47. The method of claim 43, wherein the chimeric 11-1F4 antibody is
administered once a month.
48. The method of claim 43, wherein the chimeric 11-1F4 antibody is
administered once a week.
49. The method of claim 43, wherein the amount is effective to cause the
patient to show signs of a therapeutic response in less than 5 weeks.

50. The method of claim 43, wherein the amount is effective to cause the
patient to show signs of a therapeutic response within about a week or
less.
51. The method of claim 43, wherein the effective dose of a chimeric 11-
1F4 antibody is 500 mg/m2 or less.
52. The method of claim 43, wherein the effective dose of a chimeric 11-
1F4 antibody is about 2,200 mg.
53. The method of claim 43, wherein the effective dose of a chimeric 11-
1F4 antibody is about 1-50 mg/kg.
54. The method of claim 43 in which the amount is effective to cause the
reduction in NT-proBNP to be sustained in the patient for at least about
six months after the administration of the chimeric 11-1F4 antibody.
55. A method of treating a patient with primary light chain (AL)
amyloidosis
involving the kidneys comprising administering to said patient a dose of
a chimeric 11-1F4 antibody effective to cause at least a 40% reduction
in proteinuria for said patient following administration of the chimeric 11-
1F4 antibody compared to pre-treatment level.
56. The method of claim 54, wherein the AL amyloidosis is refractory.
57. The method of claim 55, wherein the chimeric 11-1F4 antibody
comprises a constant region is derived from a human IgG1.
58. The method of claim 55, wherein the chimeric 11-1F4 antibody is
administered once a month.
59. The method of claim 55, wherein the chimeric 11-1F4 antibody is
administered once a week.
60. The method of claim 55, wherein the dose is effective to cause the
patient to show signs of a therapeutic response in less than 5 weeks.
81

61. The method of claim 55, wherein the dose is effective to cause the
patient to show signs of a therapeutic response within about a week or
less.
62. The method of claim 55, wherein the effective dose of a chimeric 11-
1F4 antibody is 500 mg/m2 or less.
63. The method of claim 55, wherein the effective dose of a chimeric 11-
1F4 antibody is about 2,200 mg.
64. The method of claim 55, wherein the effective dose of a chimeric 11-
1F4 antibody is about 1-50 mg/kg.
65. The method of claim 55 wherein the dose is effective to cause the
reduction in proteinuria to be sustained in the patient for at least about
six months after the administration of the chimeric 11-1F4 antibody.
66. A method of decreasing the amount of kappa and/or lambda light chain
fibril aggregate deposits in a human patient with primary amyloidosis
comprising kappa or lambda light chain fibril aggregate deposits
comprising administering to said patient a therapeutically effective fibril
aggregate deposit reducing dose of an antibody comprising:
a. a V K region comprising SEQ ID NO: 47,
b. a V H region comprising SEQ ID NO: 48, and
c. a human IgG1 constant region;
wherein administration of the antibody decreases the amount of kappa
and/or lambda light chain fibril aggregate deposits in the patient.
67. The method of claim 66, wherein the primary amyloidosis consists of
lambda light chain fibril aggregate deposits.
68. The method of claim 66, wherein the primary amyloidosis consists of
kappa light chain fibril aggregate deposits.
82

69. The method of claim 66, wherein the primary amyloidosis consists of
kappa and lambda light chain fibril aggregate deposits.
70. The method of claim 66, wherein the chimeric antibody is administered
once a month.
71. The method of claim 66, wherein the chimeric antibody is administered
once a week.
72. The method of claim 66, wherein the dose is effective to cause the
patient to show signs of a therapeutic response in less than 5 weeks.
73. The method of claim 66, wherein the dose is effective to cause the
patient to show signs of a therapeutic response within about a week or
less.
74. The method of claim 66, wherein the effective dose of a chimeric
antibody is 500 mg/m2 or less.
75. The method of claim 66, wherein the effective dose of a chimeric
antibody is about 2,200 mg.
76. The method of claim 66, wherein the effective dose of a chimeric
antibody is about 1-50 mg/kg.
77. The method of claim 66, wherein the dose is effective to cause organ
dysfunction in the patient to be decreased.
78. A method of treating AL amyloidosis in a patient with AL amyloidosis
comprising administering to said patient a therapeutically effective
amount of a monoclonal antibody comprising the variable regions of an
11-1F4 antibody, wherein the antibody is not murine 11-1F4.
79. The method of claim 77, wherein the antibody is a mouse-human
chimeric antibody.
80. The method of claim 77, wherein the antibody comprises a human IgG1
constant region.
83

81. A method of improving myocardial function in a patient diagnosed with
amyloid light chain amyloidosis (ALA) with cardiac involvement comprising
administering to said patient an amount of a humanized or chimeric
antibody or an antigen-binding fragment thereof, the antibody or antigen-
binding fragment comprising:
a variable heavy chain (V H) comprising: a complementarity determining
region (CDR) H1 comprising SEQ ID NO: 53; a CDRH2 comprising
SEQ ID NO: 53; and a CDRH3 comprising SEQ ID NO: 54; and
a variable light chain (V K) comprising a CDRL1 comprising SEQ ID NO:
49; a CDRL2 comprising SEQ ID NO: 50; and a CDRL3
comprising SEQ ID NO: 51;
the amount being effective to improve the myocardial function of the
patient within about three weeks of administration of the antibody or
antigen-binding fragment thereof.
82. The method of claim 81, wherein the antibody or antigen-binding
fragment
thereof is a humanized antibody.
83. The method of claim 81, wherein the antibody or antigen-binding
fragment
thereof is a chimeric antibody.
84. The method of claim 83, wherein the V K region comprises SEQ ID NO: 47
and a V H region comprising SEQ ID NO: 48.
85. The method of claim 83, wherein the antibody comprises a constant
region is derived from a human IgG1.
86. The method of claim 81, wherein the improvement in myocardial function
persists for at least three months after administration of the antibody or
antigen-binding fragment thereof.
87. The method of claims 81, wherein the antibody or antigen-binding
fragment thereof is administered no more than once, twice, three, or four
times within a three month period.
84

88. The method of claim 81, wherein the improvement in myocardial function
comprises an improvement in global longitudinal strain (GLS) compared
with pretreatment GLS level.
89. The method of claim 81, wherein the patient exhibits a pretreatment NT-
proBNP level greater than 650 pg/mL.
90. The method of claim 89, wherein the amount is effective to cause the
patient to exhibits a reduction in posttreatment NT-proBNP level of about
300 pg/mL or more compared to pretreatment NT-proBNP level.
91. The method of claim 89, wherein the improvement in myocardial function
comprises a reduction in posttreatment NT-proBNP level of about 30% or
more compared to pretreatment NT-proBNP level.
92. The method of claim 81, wherein the patient suffers from relapse or
refractory ALA.
93. The method of claim 81, wherein the ALA is further characterized as
having light chain lambda amyloid cardiac involvement.
94. The method of claim 81, wherein the ALA is further characterized as
having light chain kappa amyloid cardiac involvement.
95. The method of claim 81, wherein the ALA is not hematologically
controlled.
96. A method of monitoring improvement in myocardial function in a patient
diagnosed with light chain amyloidosis (ALA) with cardiac involvement
comprising observing an improvement in myocardial function in a patient
diagnosed with ALA having a cardiac involvement within about three
weeks after administration to said patient of a therapeutically effective
amount of a humanized or chimeric 11-1F4 antibody or an antigen-binding
fragment thereof.
97. The method of claim 96, wherein the patient suffers from relapse or
refractory ALA.

98. The method of claim 96, wherein the ALA is further characterized as
having light chain lambda amyloid cardiac involvement.
99. The method of claim 96, wherein the ALA is further characterized as
having light chain kappa amyloid cardiac involvement.
100. A method of treating amyloid light chain amyloidosis (ALA) with cardiac
involvement in a patient, wherein the ALA is not hematologically
controlled, the method comprising, administering to said patient with ALA
characterized by cardiac involvement and a lack of hematological control a
therapeutically effective amount of a humanized or chimeric antibody or an
antigen-binding fragment thereof comprising:
a variable heavy chain (V H) comprising: a complementarity determining
region (CDR) H1 comprising SEQ ID NO: 52; a CDRH2 comprising
SEQ ID NO: 53; and a CDRH3 comprising SEQ ID NO: 54; and
a variable light chain (V K) comprising a CDRL1 comprising SEQ ID NO:
49; a CDRL2 comprising SEQ ID NO: 50; and a CDRL3
comprising SEQ ID NO: 51.
101. The method of claim 100, wherein the antibody or antigen-binding
fragment thereof is a humanized antibody.
102. The method of claim 100, wherein the antibody or antigen-binding
fragment thereof is a chimeric antibody.
103. The method of claim 102, wherein the V K region comprising SEQ ID NO:
47 and a V H region comprising SEQ ID NO: 48.
104. The method of claim 102, wherein the antibody comprises a constant
region is derived from a human IgG1.
105. The method of claim 100 further comprising administering a
chemotherapeutic compound to the patient.
106. The method of claim 100, wherein the lack of hematological control
comprises circulating levels of amyloid precursor protein of at least,
86

wherein the difference between involved and uninvolved free light chains
in the subject's serum is >40mg/L.
107. The method of claim 100, wherein the patient exhibits an improvement in
global longitudinal strain (GLS) compared with pretreatment GLS level.
108. The method of claim 107, wherein the improvement in GLS occurs within
three weeks of being administered the antibody or antigen-binding
fragment thereof.
109. The method of claim 107, wherein the improvement in GLS persists for at
least three months following administration of the antibody or antigen-
binding fragment thereof.
110. The method of claim 100, wherein the ALA is further characterized as
having light chain lambda amyloid cardiac involvement.
111. The method of claim 100, wherein the ALA is further characterized as
having light chain kappa amyloid cardiac involvement
87

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
METHODS AND COMPOSITIONS FOR TREATMENT
OF AMYLOID DEPOSITION DISEASES
REFERENCE TO A SEQUENCE LISTING SUBMITTED BY EFS-WEB
[0001] The contents of the ASCII text file of the sequence listing named
8441-0009-1-5T25", which is 17.4 kb in size, was created on June 15, 2018,
and was electronically submitted via EFS-Web with this application, is
incorporated herein by reference in its entirety.
GOVERNMENT RIGHTS
[0002] This invention was made with United States government support
under grant FD-U-005110, awarded by the Food and Drug Administration.
Thus, the United States government may have certain rights to the invention
described and claimed herein.
CLAIM FOR PRIORITY
[0003] This application claims priority from United States provisional
patent application 62/539,821, filed August 1, 2017, and from United States
provisional patent application 62/637,609, filed March 2, 2018, both of which
are incorporated herein by reference in their entirety.
FIELD OF THE INVENTION
[0004] The present disclosure relates to humanized and chimeric (e.g.,
mouse-human) antibodies and antigen-binding fragments thereof useful to treat
amyloid deposition diseases, particularly primary (AL) amyloidosis,
pharmaceutical compositions comprising such antibodies, and methods of
treating amyloid deposition diseases using said antibodies and pharmaceutical
compositions. The disclosure additionally relates to methods of treating
amyloid
deposition diseases with amyloid fibril-reactive antibodies. In particular,
the
present disclosure relates to methods of improving myocardial function in
patients diagnosed with amyloid light chain amyloidosis (ALA) that includes
cardiac involvement (i.e., amyloid deposition in or around the heart). Such
patients may have ALA deposits comprising light chain lambda amyloid or light
1

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
chain kappa amyloid. Moreover, the patients may have a disease that is
hematologically controlled or uncontrolled.
BACKGROUND OF THE INVENTION
[0005] The following discussion is provided merely to aid the reader in
understanding the disclosure and is not admitted to describe or constitute
prior
art thereto.
[0006] Native antibodies are usually heterotetrameric glycoproteins of
about 150,000 daltons composed of two identical light chains and two identical

heavy chains. Each light chain is linked to a heavy chain by one disulfide
bond, while the number of additional disulfide linkages between the heavy
chains varies with different antibody isotypes. The simplest isotype is IgG,
which comprises just two light chains and two heavy chains, in which the two
heavy chains are linked by two disulfide linkages. Each heavy chain has a
variable domain (VH) at one end with a number of adjacent constant domains.
Each light chain has a variable domain (VL) at one end and a constant domain
at its other end. Each variable domain of the light and heavy chain in an
antibody comprises three segments called complementarity-determining
regions ("CDR") or hypervariable regions. Each CDR in a light chain, together
with the corresponding CDR in the adjacent heavy chain, form an antigen-
binding site of the antibody. Light chains are of two major types, K and A,
depending on their constant region. Both K and A light chains may combine
with any of the different heavy chain types.
[0007] Amyloid light-chain amyloidosis (AL amyloidosis, AL, or ALA),
also called primary amyloidosis, is the most common form of systemic
amyloidosis in the United States. The term "amyloidosis" refers to a cluster
of
diseases which share a common feature, i.e., the extracellular deposition of
pathologic insoluble fibrillar proteins in organs and tissues (Rodney, et al.
¨
NEJM, 25:898). Amyloidosis is caused by malfunction of a person's antibody-
producing cells causing production of abnormal protein fibers which aggregate
to form insoluble amyloid deposits in organs and tissues. The type of
2

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
amyloidosis is determined by the nature of the precursor proteins which form
the fibril deposit. In primary amyloidosis, the fibrils comprise fragments of
immunoglobulin light chains and in secondary amyloidosis, the fibrils comprise

amyloid A protein. Modern classification of amyloidosis is based on the nature

of the precursor plasma proteins which form the fibril deposit.
[0008] The
precursor plasma proteins are diverse and unrelated.
Nevertheless, all precursor deposits produce amyloid deposits that share a
common typical 8-pleated-sheet configuration, which is responsible for the
typical staining properties of the fibrillar deposits. The final stage in the
development of amyloidosis is the deposit of amyloid fibrils in the organs of
the
sufferer. Amyloidosis mortality is high, with current five-year survival rates
of
about 28%.
[0009] To date,
the treatment of AL has been directed towards reducing
the synthesis of amyloidogenic precursor light chains by attacking the
malfunctioning cells through conventional or high dose cytotoxic
chemotherapy. This treatment suffers from two disadvantages. First, the
fibrillar deposits are often asymptomatic until after significant deposition
has
taken place.
Therefore, treatment is unlikely to be undertaken before
significant deposits have already occurred. Second, since this treatment is,
at
best, effective only to stop the production of precursor abnormal protein but
not to remove the existing deposits, prognosis for AL patients remains
exceedingly poor due to persistence (or progression) of the pathologic
deposits (Solomon, et al. ¨ Int. J. Exp. Clin. Invest. 2:269)
[0010] As a
result, therapeutic targeting and clearance of amyloid
deposits is an area of intense medical interest. However, the FDA has not
approved any such therapeutic products to date, and therefore, there is still
a
significant unmet medical need. The compositions and methods disclosed
herein fulfill this need.
3

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
SUMMARY OF THE INVENTION
[0011] Described herein are compositions and methods for treating
amyloid deposition diseases, specifically primary (AL) amyloidosis. The
disclosed compositions and methods employ humanized or chimeric
antibodies or fragments thereof that specifically bind to amyloid fibrils
(e.g.,
amyloid light chain fibrils) to target the fibrils for clearance by the immune

system.
[0012] In one aspect, the present compositions and methods comprise
humanized or chimeric antibodies (e.g., mouse-human chimeric antibodies)
useful for treatment of amyloid deposition diseases, particularly primary
(ALA)
amyloidosis. In some embodiments, the disclosed antibody comprises a VK
region comprising SEQ ID NO: 47 and a VH region comprising SEQ ID NO: 48.
In some embodiments, the antibody comprises a constant region derived from
a human IgG1. In some embodiments, the antibody binds to amyloid fibrils
with a higher affinity than its murine equivalent. In some embodiments, the
antibody binds to an epitope expressed by the 8-pleated sheet configuration of

amyloid fibrils with higher affinity than a mouse antibody comprising a VK
region of SEQ ID NO: 36 and a VH region of SEQ ID NO: 35. And in some
embodiments, the antibody binds to kappa and lambda amyloid fibrils in vivo.
[0013] In another aspect, the present disclosure provides
pharmaceutical compositions comprising the disclosed humanized or chimeric
antibody and a pharmaceutically acceptable carrier.
[0014] A chimeric antibody useful in the subject methods and
pharmaceutical compositions may be produced by co-transfection in
mammalian cells of the vector constructs 11-1F4VK.pKN100 and 11-
F4VH.pG1D200 or transfection in mammalian cells of the supervector
construct pG1KD200-11-1F4. In some embodiments, the co-transfection of
the vector constructs 11-1F4VK.pKN100 and 11-F4VH.pG1D200 or
transfection of a supervector construct pG1KD200-11-1F4 takes place in COS
cells. The antibody produced is designated "chimeric 11-1F4 antibody".
4

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[0015] In another aspect, the present disclosure provides methods for
treating or ameliorating amyloid deposition diseases, such as primary (AL)
amyloidosis, in a human in need of such treatment by administering to a
human patient in need of such treatment or amelioration a therapeutically
effective amount of at least one of the disclosed antibodies or fragments in
an
amount effective to treat or ameliorate the amyloid deposition disease and/or
the symptoms of the disease. In some embodiments, the disclosed antibody
may be administered together with a pharmaceutically acceptable carrier.
[0016] In some embodiments, the amyloid deposition disease is primary
amyloidosis. In some embodiments, the antibody is administered in a dose of
about 500 mg/m2 or less, while in some embodiments, the effective dose of a
chimeric 11-1F4 antibody is about 2,200 mg, and in some embodiments, the
effective amount is about 1-50 mg/kg.
[0017] In some embodiments, the primary amyloidosis comprises
involvement of at least one organ or tissue selected from the group consisting

of heart, kidneys, liver, lung, gastrointestinal tract, nervous system,
muscular
skeletal system, soft tissue, and skin.
[0018] In some embodiments when the amyloidosis is effecting the
heart, the patient's N-terminal pro b-type natriuretic peptide (NT-proBNP)
level
may decrease following administration of the antibody by at least about 30% or

about 40% compared to baseline levels. In some embodiments, the patient's
N-terminal pro b-type natriuretic peptide (NT-proBNP) level may decrease to
less than about 9100, about 8000, about 7000, about 6000, or about 5000
ng/L following administration of the antibody. In some embodiments, the
patient was classified as New York Heart Association (NYHA) Functional
Classification class II or III prior to administration of the antibody and is
classified as class I following administration of the antibody.
[0019] In some embodiments when the amyloidosis is affecting the
kidneys, the patient's urine protein level may decrease by at least about 30%
or about 40% compared to baseline levels following administration of the

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
antibody. In some embodiments, the patient's urine protein may decrease to
less than about 7000, about 6000, about 5000, or about 4000 mg/24 hours
following administration of the antibody.
[0020] In some embodiments, administration of the antibody does not
cause any serious adverse events.
[0021] The present disclosure provides methods of treating ALA with
cardiac involvement. The disclosed methods are uniquely able to provide
beneficial clinical results within about 3 weeks of the commencement of
treatment and in a patient population that was previously considered incapable
of
treating due to an extremely short life expectancy.
[0022] In one aspect, the present disclosure provides methods of
improving myocardial function in a patient diagnosed with amyloid light chain
amyloidosis (ALA) with cardiac involvement comprising: administering to a
patient diagnosed with ALA with cardiac involvement a therapeutically
effective
amount of a humanized or chimeric antibody or an antigen-binding fragment
thereof, the antibody or antigen-binding fragment comprising: a variable heavy

chain (VH) comprising: a complementarity determining region (CDR) H1
comprising SEQ ID NO: 52; a CDRH2 comprising SEQ ID NO: 53; and a CDRH3
comprising SEQ ID NO: 54; and a variable light chain (VK) comprising a CDRL1
comprising SEQ ID NO: 49; a CDRL2 comprising SEQ ID NO: 50; and a CDRL3
comprising SEQ ID NO: Si; thereby improving the myocardial function of the
patient within about three weeks of administration of the antibody or an
antigen-
binding fragment thereof.
[0023] In another aspect, the present disclosure provides methods of
monitoring improvement in myocardial function in a patient diagnosed with
light
chain amyloidosis (ALA) with cardiac involvement comprising observing an
improvement in myocardial function in a patient diagnosed with ALA having a
cardiac involvement within about three weeks after administration to said
patient
of a therapeutically effective amount of a humanized or chimeric 11-1F4
antibody
or an antigen-binding fragment thereof.
6

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[0024] In another aspect, the present disclosure provides methods of
treating amyloid light chain amyloidosis (ALA) with cardiac involvement in a
patient, wherein the ALA is not hematologically controlled, the method
comprising administering to a patient with ALA characterized by cardiac
involvement and a lack of hematological control a therapeutically effective
amount of a humanized or chimeric antibody or an antigen-binding fragment
thereof comprising: a variable heavy chain (VH) comprising: a complementarity
determining region (CDR) H1 comprising SEQ ID NO: 52; a CDRH2 comprising
SEQ ID NO: 53; and a CDRH3 comprising SEQ ID NO: 54; and a variable light
chain (VK) comprising a CDRL1 comprising SEQ ID NO: 49; a CDRL2 comprising
SEQ ID NO: 50; and a CDRL3 comprising SEQ ID NO: 51.
[0025] In some embodiments of the foregoing aspects, the antibody or
antigen-binding fragment thereof may be a humanized antibody, while in some
embodiments, the antibody or antigen-binding fragment thereof may be a
chimeric antibody.
[0026] In some embodiments of the foregoing aspects, the VK region of the

antibody or antigen-binding fragment may comprise SEQ ID NO: 47 and the VH
region may comprise SEQ ID NO: 48.
[0027] In some embodiments of the foregoing aspects, the antibody or
antigen-binding fragment may comprise a constant region that is derived from a

human IgG1.
[0028] In some embodiments of the foregoing aspects, the methods may
provide improvement in myocardial function that persists for at least three
months after administration of the antibody or antigen-binding fragment
thereof.
In some embodiments, the improvement may persist for four, five, six, seven,
eight, nine, ten, eleven, or twelve or more months.
[0029] In some embodiments of the foregoing aspects, the antibody or
antigen-binding fragment thereof may be administered no more than once, twice,

three, or four times within a three month period. In some embodiments, the
7

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
antibody or antigen-being fragment may be administered even less, for example,

once every other month or once every three months.
[0030] In some
embodiments of the foregoing aspects, the methods
provide an improvement in myocardial function that may comprise an
improvement in global longitudinal strain (GLS) compared with pretreatment GLS

level.
[0031] In some
embodiments of the foregoing aspects, the patient exhibits
a pretreatment NT-proBNP level greater than 650 pg/mL.
[0032] In some
embodiments of the foregoing aspects, the therapeutically
effective dose or amount of the antibody or antibody fragment is effective to
cause a reduction in the patient's post-treatment NT-proBNP level of about 300
pg/mL or more compared to pretreatment NT-proBNP level. In some
embodiments, the reduction in NT-proBNP may be about 400, about 500, about
600, about 700, about 800, about 900, or about 1000 or more pg/mL.
[0033] In some
embodiments of the foregoing aspects, the methods
provide an improvement in myocardial function that may comprise a reduction in

post-treatment NT-proBNP level of about 30% or more compared to pretreatment
NT-proBNP level.
[0034] In some
embodiments of the foregoing aspects, the patient may
suffer from relapse or refractory ALA.
[0035] In some
embodiments of the foregoing aspects, the ALA may be
further characterized as having light chain lambda amyloid cardiac
involvement,
while in some embodiments, the ALA may be further characterized as having
light chain kappa amyloid cardiac involvement.
[0036] In some
embodiments of the foregoing aspects, the ALA may not
be hematologically controlled. For example, the difference between involved
and
uninvolved free light chains in the subject's serum may be >40mg/L or the
subject may have detectable levels of toxic amyloid precursor proteins in his
or
her blood or serum.
8

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[0037] In some embodiments of the foregoing aspects, the methods may
further comprise administering a chemotherapeutic compound to the patient.
[0038] In another aspect, the present disclosure provides methods of
detection of an amyloid deposition disease in a patient suspected of having
such disease by administering a labeled antibody or an antigen-binding
fragment thereof and detecting the presence of the label in the patient. In
some embodiments, the label may be a radiolabel, such as 1241, but other sorts

of labels can be readily envisioned by one of skill in the art.
[0039] In another aspect, the present disclosure provides methods of
treating a patient with an amyloid deposit disease comprising administering a
therapeutically effective amount of a humanized or chimeric 11-1F4 antibody
or an antigen-binding fragment thereof to said patient less frequently than
once per month. For instance, in some embodiments, treatment may require
that the patient is administered a therapeutically effective amount of the
humanized or chimeric 11-1F4 antibody or antigen-binding fragment only once
every two months, once every three months, once every four months, once
every five months, once every six months, once every seven months, once
every eight months, once every nine months, once every ten months, once
every eleven months, or once a year.
[0040] In some embodiments of this aspect, the humanized or chimeric
11-1F4 antibody comprises a constant region is derived from a human IgG1.
[0041] In some embodiments of this aspect, the amyloid deposition
disease is primary light chain (AL) amyloidosis, and the disease may comprise
aggregates of lambda light chain fibrils. In some embodiments, the presence
of aggregates of lambda light chain fibrils is significantly reduced following

administration of the antibody.
[0042] In some embodiments of this aspect, the therapeutically effective
amount of the humanized or chimeric 11-1F4 antibody or antigen-binding
fragment is 500 mg/m2 or less, while in some embodiments, the therapeutically
effective amount of the humanized or chimeric 11-1F4 antibody or antigen-
9

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
binding antibody fragment is about 2,200 mg, and in some embodiments, the
therapeutically effective amount is about 1-50 mg/kg.
[0043] In some embodiments of this aspect, the therapeutically effective
amount of the humanized or chimeric 11-1F4 mAb or antigen-binding fragment
is administered once every two, three, four, five, or six months. In some
embodiments, the therapeutically effective amount of the chimeric 11-1F4 mAb
or antigen-binding fragment is administered biannually or only once a year.
[0044] In another aspect, the present disclosure provides methods of
treating a patient with primary light chain (AL) amyloidosis involving the
heart
comprising administering a dose of a humanized or chimeric 11-1F4 antibody
to said patient, said dose being effective to cause at least a 30% reduction
in
N-terminal pro b-type natriuretic peptide (NT-proBNP) level in said patient
following administration of the chimeric 11-1F4 antibody compared to pre-
treatment level. In some embodiments, the AL amyloidosis is refractory.
[0045] In some embodiments of this aspect, the humanized or chimeric
11-1F4 antibody comprises a constant region is derived from a human IgGl.
[0046] In some embodiments of this aspect, the humanized or chimeric
11-1F4 antibody is administered once a month, while in some embodiments,
the humanized or chimeric 11-1F4 antibody is administered once a week.
[0047] In some embodiments of this aspect, the therapeutically effective
amount of the humanized or chimeric 11-1F4 antibody is 500 mg/m2 or less,
while in some embodiments, the therapeutically effective amount of the
humanized or chimeric 11-1F4 antibody is about 2,200 mg, and in some
embodiments, the effective amount is about 1-50 mg/kg.
[0048] In some embodiments of this aspect, the reduction in NT-proBNP
is sustained in the patient for at least about six months after the
administration
of the chimeric 11-1F4 antibody.
[0049] In another aspect, the present disclosure provides methods of
treating a patient with primary light chain (AL) amyloidosis involving the

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
kidneys comprising administering a dose of a humanized or chimeric 11-1F4
antibody or antigen-binding antibody fragment to said patient, said dose being

effective to cause at least a 40% reduction in proteinuria in said patient
following administration of the humanized or chimeric 11-1F4 antibody
compared to pre-treatment level. In some embodiments, the AL amyloidosis is
refractory.
[0050] In some embodiments of this aspect, the humanized or chimeric
11-1F4 antibody comprises a constant region is derived from a human IgG1.
[0051] In some embodiments of this aspect, the humanized or chimeric
11-1F4 antibody is administered once a month, while in some embodiments,
the humanized or chimeric 11-1F4 antibody is administered once a week.
[0052] In some embodiments of this aspect, the therapeutically effective
amount of the humanized or chimeric 11-1F4 antibody is 500 mg/m2 or less,
while in some embodiments, the therapeutically effective amount of the
humanized or chimeric 11-1F4 antibody is about 2,200 mg, and in some
embodiments, the therapeutically effective amount is about 1-50 mg/kg.
[0053] In some embodiments of this aspect, the reduction in proteinuria
is sustained in the patient for at least about six months after the
administration
of the chimeric 11-1F4 antibody.
[0054] In another aspect, the present disclosure provides methods of
decreasing the amount of kappa or lambda light chain fibril aggregate deposits

in a patient in need thereof comprising, administering to said patient a dose
of
an antibody comprising: (a) a VK region comprising SEQ ID NO: 47, (b) a VH
region comprising SEQ ID NO: 48, and (c) a human IgG1 constant region; said
dose being effective to decrease the amount of kappa or lambda light chain
fibril aggregate deposits in the patient.
[0055] In some embodiments of this aspect, the primary amyloidosis
consists of lambda light chain fibril aggregate deposits, while in some
embodiments, the primary amyloidosis consists of kappa light chain fibril
11

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
aggregate deposits, and in still other embodiments, the primary amyloidosis
consists of kappa and lambda light chain fibril aggregate deposits.
[0056] In some embodiments of this aspect, the humanized or chimeric
11-1F4 antibody is administered once a month, while in some embodiments,
the humanized or chimeric 11-1F4 antibody is administered once a week.
[0057] In some embodiments of this aspect, the therapeutically effective
amount of the humanized or chimeric 11-1F4 antibody is 500 mg/m2 or less,
while in some embodiments, the therapeutically effective amount of the
humanized or chimeric 11-1F4 antibody is about 2,200 mg, and in some
embodiments, the therapeutically effective amount is about 1-50 mg/kg.
[0058] In another aspect, the present disclosure provide methods of
treating AL amyloidosis comprising administering to a patient with AL
amyloidosis a monoclonal comprising the complementarity determining
regions (CDRs) of an 11-1F4 antibody, wherein the antibody is not murine 11-
14F.
[0059] In some embodiments of this aspect, the antibody may be a
mouse-human chimeric antibody.
[0060] In some embodiments of this aspect, the antibody comprises a
human IgG1 constant region.
[0061] In some embodiments of any of the foregoing methods, organ
dysfunction in the patient is decreased following administration of the
antibody.
In some embodiments of any of the foregoing methods, the patient shows
signs of a therapeutic response in less than 5 weeks after treatment,
sometimes in less than 4 weeks after treatment, sometimes in less than 3
weeks after treatment, sometimes in less than 2 weeks after treatment, while
in some other embodiments, the patient shows signs of a therapeutic response
within about a week or less after treatment.
12

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[0062] The foregoing general description and following detailed
description are exemplary and explanatory and are intended to provide further
explanation of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS
[0063] Figure 1 outlines the strategy used to clone the murine VH and
VK genes from a hybridoma cell line.
[0064] Figure 2 is a listing of DNA and amino acid sequences of the
murine 11-1F4 antibody VH region gene, SEQ ID NO: 39 and NO: 35,
respectively.
[0065] Figure 3 is a listing of DNA and amino acid sequences of the
murine 11-1F4 antibody VK region gene, SEQ ID NO: 40 and NO: 36,
respectively.
[0066] Figure 4 is a map of the immunoglobulin kappa light chain
expression vector pKN100. It consists of a pSV2 vector fragment, which has
the SV40 early and crippled SV40 late promoter, the SV40 origin and the
Col El origin. It also has the ampicillin resistance and neo genes. The
crippled
SV40 late promoter drives the neo genes. It also has the HCMVi promoter, a
multiple cloning site (containing the BamHI and HindlIl restriction sites) for
the
insertion of an immunoglobulin variable region gene, and cDNA for the human
kappa constant region gene terminated by a spaC2 termination signal
sequence ("Arnie"), which is in the same orientation as the kappa light chain
expression cassette.
[0067] Figure 5 is a map of the immunoglobulin gamma 1 heavy chain
expression vector pG1D200. It consists of a pSV2dhfr vector fragment, which
has the SV40 early and crippled SV40 late promoter, the SV40 origin, and the
Col El origin. It also has the ampicillin resistance and dhfr genes. The
crippled
SV40 late promoter drives the dhfr gene. Consequently, expression is poor,
allowing for the selection of multigene/high expression level clones using
comparatively low levels of methotrexate. It also has the HCMVi promoter
13

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
fragment, a multiple cloning site, cDNA for a human gamma 1 constant region
gene (intron minus) which is followed by a spaC2 termination signal sequence
("Arnie").
[0068] Figure 6 is a listing of the DNA and amino acid sequences of the
modified murine 11-1F4 antibody VK region gene (SEQ ID NO: 42 and NO: 47,
respectively) and the sequences of the oligonucleotide primers used to modify
the VK gene (SEQ ID NO: 41 and NO: 43, respectively).
[0069] Figure 7 is a listing of the DNA and amino acid sequences of the
modified murine 11-1F4 antibody VH region gene (SEQ ID NO: 45 and NO: 48,
respectively) and the sequences of the oligonucleotide primers used to modify
the VH gene (SEQ ID NO: 44 and NO: 46, respectively).
[0070] Figure 8 is a graphical representation of the result of the
amyloid
fibril binding ELISA assay. The cos cell supernatants containing chimeric 11-
1F4 antibody were tested separately on the same ELISA plate along with
purified murine 11-1F4 antibody. The absorbance was read at 0D405. New
sv = pG1KD200-11-1F4. New co-transfection = 11-1F4VHpG1D200 plus 11-
1F4VK. pKN100.
[0071] Figure 9 shows clearance of human ALk and human ALA
amyloidomas in mice treated with murine 11-1F4. Mice were treated with
either a single dose (Panel A) or multiple doses (Panel B) of murine 11-1F4.
The results indicate that murine 11-1F4 quickly clears ALk amyloidomas, but in

most instances multiple doses were required to clear ALA amyloidomas from
the mice.
[0072] Figure 10 shows the dosing/evaluation scheme of Phase 1a/b
trials of chimeric 11-1F4. Panel A shows the scheme for Phase la and Panel
B shows the scheme for Phase lb. Panel C shows the doses that were used
in these studies.
[0073] Figure 11 shows that administration of chimeric 11-1F4 provides
an improvement in cardiac function in most patients. Panel A shows the
14

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
results from a Phase 1a/b trial in a bar graph, and Panel B shows the results
as a box plot.
[0074] Figure 12 shows cardiac response (NT-proBNP) in an exemplary
patient during Phase 1a/b clinical trial of c11-1F4 antibody.
[0075] Figure 13 shows that administration of chimeric 11-1F4 provides
an improvement in renal function in most patients. Panel A shows the results
from a Phase la/b trial in a bar graph, and Panel B shows the results as a box

plot.
[0076] Figure 14 shows renal response (proteinuria) in an exemplary
patient during Phase 1a/b clinical trial of chimeric 11-1F4 antibody.
[0077] Figure 15 shows a graphical depiction of changes in global
longitudinal strain with the chimeric 11-1F4 monoclonal antibody.
[0078] Figure 16 shows a graphical depiction of organ response (NT-
proBNP) upon chimeric 11-1F4 antibody treatment in a patient who had a partial

hematological response to chemotherapy with no organ response prior to the
chimeric antibody treatment.
[0079] Figure 17 shows an echocardiogram of an amyloidosis patient with
cardiac involvement at week 0 and at week 12 post chimeric 11-1F4 mAb
treatment.
DETAILED DESCRIPTION OF THE INVENTION
[0080] In accordance with the present disclosure, methods and
compositions comprising humanized antibodies, chimeric antibodies (e.g.,
mouse-human antibodies) or antigen-binding fragments thereof are provided that

are useful for administration to humans suffering from amyloid deposition
diseases to treat or ameliorate the disease and symptoms of the disease. The
antibodies and antibody fragments of the invention bind to amyloid deposits
and
activate the patient's immune system to clear the bound materials while
producing little or no human anti-mouse antibody (HAMA) reaction. The
disclosure provides pharmaceutical compositions comprising at least one of
said

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
antibodies or antibody fragments and a pharmaceutically acceptable carrier and

methods of treating or ameliorating the amyloidosis and the symptoms of
amyloidosis by administering to a patent an amount of said antibody or
antibody
fragment effective to remove at least some of the amyloid deposits from the
patient's organs and thus to treat or ameliorate the disease and its symptoms.
[0081] Further in accordance with the present disclosure, methods are
provided for treating amyloid deposition diseases. In particular, the present
disclosure is directed towards improving myocardial function in a patient
diagnosed with amyloid light chain amyloidosis (ALA) that has cardiac
involvement. The methods comprise administering to a patient a humanized or
chimeric antibody (e.g., mouse-human antibody) or an antigen-binding fragment
thereof that binds to amyloid fibril deposits, circulating amyloids, and toxic

amyloid precursor protein. In particular, the disclosure shows that
administering
the disclosed amyloid fibril-binding antibodies to patients diagnosed with ALA

with cardiac involvement results in improved global longitudinal strain (GLS)
compared to pretreatment GLS level, and/or a reduction in NT-proBNP level
compared to pretreatment NT-proBNP level. Additionally, patients may be
effectively treated even if their disease is not hematologically controlled
(i.e., the
patient has detectable levels of toxic amyloid precursor proteins in
circulation or
when the difference between involved and uninvolved free light chains of >40
mg/L) and regardless of whether the disease involves fibrils of kappa or
lambda
proteins.
Definitions
[0082] It is to be understood that methods are not limited to the
particular embodiments described, and as such may vary. It is also to be
understood that the terminology used herein is for the purpose of describing
particular embodiments only, and is not intended to be limiting. The scope of
the present technology will be limited only by the appended claims.
[0083] As used herein, certain terms may have the following defined
meanings. As used in the specification and claims, the singular form "a," "an"
16

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
and "the" include singular and plural references unless the context clearly
dictates otherwise. For example, the term "a cell" includes a single cell as
well
as a plurality of cells, including mixtures thereof.
[0084] As used herein, the term "comprising" is intended to mean that
the compositions and methods include the recited elements, but not excluding
others. "Consisting essentially of" when used to define compositions and
methods, shall mean excluding other elements of any essential significance to
the composition or method. "Consisting of" shall mean excluding more than
trace elements of other ingredients for claimed compositions and substantial
method steps. Embodiments defined by each of these transition terms are
within the scope of this disclosure. Accordingly, it is intended that the
methods
and compositions can include additional steps and components (comprising)
or alternatively including steps and compositions of no significance
(consisting
essentially of) or alternatively, intending only the stated method steps or
compositions (consisting of).
[0085] As used herein, "about" means plus or minus 10%.
[0086] As used herein, "optional" or "optionally" means that the
subsequently described event or circumstance may or may not occur, and that
the description includes instances where said event or circumstance occurs
and instances where it does not.
[0087] As used herein, the terms "individual", "patient", or "subject"
can
be an individual organism, a vertebrate, a mammal (e.g., a bovine, a canine, a

feline, or an equine), or a human. In a preferred embodiment, the individual,
patient, or subject is a human.
[0088] As used herein, the term an "isolated antibody" is intended to
refer to an antibody which is substantially free of other antibodies having
different antigenic specificities (e.g., an isolated antibody that
specifically binds
to an amyloid fibril is substantially free of antibodies that do not bind to
amyloid
fibrils). An isolated antibody that specifically binds to an epitope of an
amyloid
light chain fibril (e.g., a kappa and/or lambda fibril) may, however, have
cross-
17

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
reactivity to other proteins, such as amyloid A fibrils. However, the antibody

preferably always binds to human amyloid light chain fibrils. In addition, an
isolated antibody is typically substantially free of other cellular material
and/or
chemicals.
[0089] As used
herein, the phrases "therapeutically effective amount"
and "therapeutic level" mean an antibody dose or plasma concentration in a
subject, respectively, that provides the specific pharmacological effect for
which the antibody is administered in a subject in need of such treatment,
i.e.,
to reduce, ameliorate, or eliminate the effects or symptoms of an amyloid
deposition disease, such as AL amyloidosis. It is
emphasized that a
therapeutically effective amount or therapeutic level of a drug will not
always
be effective in treating the conditions/diseases described herein, even though

such dosage is deemed to be a therapeutically effective amount by those of
skill in the art. The therapeutically effective amount may vary based on the
route of administration and dosage form, the age and weight of the subject,
and/or the subject's condition, including the type and stage of the
amyloidosis
at the time that treatment commences, among other factors.
[0090] The terms
"treatment" or "treating" as used herein with reference
to amyloid diseases, such as AL amyloidosis, refer to reducing, ameliorating
or
eliminating one or more symptoms or effects of the amyloidosis, including but
not limited to clearance or degradation of amyloid plaques or deposits,
improving organ function of organs effected by the disease (e.g., the heart,
kidney, liver, etc.), and increasing the patient's lifespan or 5-year
survival.
[0091] A
"therapeutic response" mean an improvement in at least one
measure of amyloid disease, such as a reduction in the size of existing
amyloid deposits or plaques, a decrease in the rate of amyloid deposition, or
improved organ function as measured by standard techniques. For instance,
in patients with amyloid deposits in the heart, improved organ function (i.e.,
a
therapeutic response) may be indicated by a decrease in the level of the
patient's N-terminal pro b-type natriuretic peptide (NT-proBNP) or a decrease
18

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
in the patient's New York Heart Association (NYHA) Functional Classification
level. In patients with amyloid deposits in the kidneys, improved organ
function (i.e., a therapeutic response) may be indicated by a decrease in
proteinuria or the rate of protein output in the urine.
[0092] As used
herein, the term "humanized antibody" refers to an
antibody that comprises the CDRs of antibodies derived from mammals other
than human, and the framework region (FR) and the constant region of a human
antibody. A humanized antibody is useful as a therapeutically effective
component in a therapeutic agent according to the present disclosure since
antigenicity of the humanized antibody in human body is lowered.
[0093] As used
herein, "cardiac involvement" means that a patient
suffering from an amyloid disease have amyloid deposits in the heart. Amyloid
deposits in the heart result in release of NT-proBNP and increased NT-proBNP
levels in the blood of the patient. Herein, a patient has cardiac involvement
if NT-
proBNP is greater than 650 pg/m L.
[0094] As used
herein, the description of not hematologically controlled"
with respect to AL amyloidosis means that the disease is not in either
complete
remission or very good partial remission. For example, the disease is not
hematologically controlled when the patient has detectable levels of toxic
amyloid
precursor proteins in circulation (i.e., blood or serum) or when the
difference
between involved and uninvolved free light chains is >40 mg/L in the patient's

blood or serum.
[0095] As used
herein, the term "serious adverse event" means an
untoward medical event that results in death, is life-threatening, requires
inpatient
hospitalization or prolongation of existing hospitalization, or results in
persistent
or significant disfigurement or disability, as defined in 21 CFR 312.32(a).
[0096] As used
herein, the term "pharmaceutically-acceptable carrier"
means a material for admixture with a pharmaceutical compound (e.g., a
chimeric antibody) for administration to a patient as described, for example,
in
19

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
"Ansel's Pharmaceutical Dosage Forms and Delivery Systems", Tenth Edition
(2014).
Anti-AL Antibodies
Murine anti-fibril antibodies
[0097] Recent animal studies have shown that the administration of the
murine 11-1F4 antibody and other murine anti-human light chain specific
antibodies directed against an epitope common to the 8-pleated-sheet structure

present on AL fibrils results in complete degradation of the human ALk and ALA

amyloid deposits. Some of these murine antibodies are described in United
States patent 8,105,594 ("the '594 patent"), which is incorporated herein by
reference in its entirety.
[0098] Murine antibodies are generally unsuitable for administration to
other animal species (such humans) because the receiving species will
recognize the murine antibody as antigenic and will produce antibodies against

it. The antigenicity of an antibody from one species when injected into
another
species is normally caused by a portion of a constant domain. Such an
antigenic response will impede or prevent the desired therapeutic effect of
the
murine antibody. In humans, this antigenic response is called human anti-
mouse antibody (HAMA). The antibodies described in the '594 patent have
the potential to be highly immunogenic in humans via the human anti-mouse
antibody (HAMA) response. Since the HAMA response usually results in the
rapid clearance of a mouse antibody from the human recipient, HAMA would
severely limit any potential human therapeutic benefit a murine antibody could

have. Therefore, these murine antibodies are unsuitable for administration to
a
patient to halt or reverse the deposition of amyloid fibrils in a patient.
Thus,
the present disclosure provides compositions and methods for treating amyloid
deposition diseases that is less likely to produce an immunogenic HAMA
response in a patient following administration.
Humanized and chimeric anti-fibril antibodies

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[0099] The
present disclosure provides humanized and chimeric
antibodies or antigen-binding fragments thereof for treating amyloidosis.
Typically, an antibody consists of four polypeptides: two identical copies of
a
heavy (H) chain polypeptide and two copies of a light (L) chain polypeptide.
Typically, each heavy chain contains one N-terminal variable (VH) region and
three C-terminal constant (CH1, CH2 and CH3) regions, and each light chain
contains one N-terminal variable (VL or VK) region and one C-terminal constant

(CL) region. Each variable domain of the light and heavy chain in an antibody
also comprises three segments called complementarity-determining regions
("CDR") or hypervariable regions. Each CDR in a light chain, together with the

corresponding CDR in the adjacent heavy chain, form an antigen-binding site of

the antibody. The variable regions of each pair of light and heavy chains form

the antigen binding site of an antibody, whereas the constant region provides
structural support and modulates the immune response initiated by the antigen
binding.
[00100]
Chimeric antibodies incorporate the variable region of a non-human
antibody into the constant region of a human antibody. A chimeric 11-1F4, for
instance, may be created by expressing the murine variable region with the Fc
region of a human antibody, such as a human IgG1.
[00101]
Humanized forms of non-human (e.g., murine) antibodies can be
obtained, which contain minimal sequences derived from non-human
immunoglobulin. In general, a humanized antibody may comprise one or two or
more variable domains in which variable regions are derived from non-human
immunoglobulin and framework regions (FR) correspond to a human
immunoglobulin sequence. Thus, in some embodiments, a humanized anti-AL
antibody comprises a human antibody framework region. Such antibodies can
be prepared by known techniques.
[00102] The
murine 11-1F4 monoclonal antibody is an anti-AL antibody
produced by the 5P2/0 hybridoma cell deposited Alan Solomon, MD (University
of Tennessee Medical Center at Knoxville, TN). The hybridoma cell line is
21

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
available from the American Type Culture Collection (ATCC access PTA-105).
The VK region (SEQ ID NO: 36) and the VH region (SEQ ID NO: 35) of the 11-
1F4 antibody are shown in Table 1 below. The CDR sequences for the heavy
and light chains and provided in Table 2.
Table 1 ¨11-1F4 monoclonal antibody variable sequences
SEQ ID NO: 36
VK region:
Asp Val Val Met Thr Gin Thr Pro Leu Ser Leu Pro Val Ser Leu Gly
Asp Gin Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Leu Val His Arg
Asn Gly Asn Thr Tyr Leu His Trp Tyr Leu Gin Lys Pro Gly Gin Ser
Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile Ser
Arg Val Glu Ala Glu Asp Leu Gly Leu Tyr Phe Cys Phe Gln Thr Thr
Tyr Val Pro Asn Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
SEQ ID NO: 35
VH region:
Gin Val Gin Leu Lys Glu Ser Gly Pro Gly Leu Val Ala Pro Ser Gin Ser
Leu Ser Ile Thr Cys Thr Val Ser Gly Phe Ser Leu Ser Ser Tyr Gly Val
Ser Trp Val Arg Gin Pro Pro Gly Lys Gly Leu Glu Trp Leu Gly Val Ile
Trp Gly Asp Gly Ser Thr Asn Tyr His Pro Asn Leu Met Ser Arg Leu
Ser Ile Ser Lys Asp Ile Ser Lys Ser Gin Val Leu Phe Lys Leu Asn Ser
Leu Gin Thr Asp Asp Thr Ala Thr Tyr Tyr Cys Val Thr Leu Asp Tyr
Trp Gly Gin Gly Thr Ser Val Thr Val Ser Ser
The complementarity determining regions (CDRs) of the variable sequences are
shown in boldface type in the table above.
Table 2 ¨11-1F4 monoclonal antibody CDR sequences
Sequence Amino Acid
CDRL1 Arg Ser Ser Gin Ser Leu Val His Arg Asn Gly Asn
(SEQ ID NO: 49) Thr Tyr Leu His
CDRL2 Lys Val Ser Asn Arg Phe Ser
(SEQ ID NO: 50)
CDRL3 Phe Gin Thr Thr Tyr Val Pro Asn Thr
(SEQ ID NO: Si)
CDRH1 Ser Tyr Gly Val Ser Trp
(SEQ ID NO: 52)
22

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
CDRH2 Val Ile Trp Gly Asp Gly Ser Thr Asn Tyr His Pro
(SEQ ID NO: 53) Asn Leu Met Ser Arg Leu Ser Ile Ser
CDRH3 Leu Asp Tyr
(SEQ ID NO: 54)
[00103] One can
clone the genes for the VH and VK regions shown above to
produce a chimeric 11-1F4 antibody using known human antibody sequences.
The chimeric 11-1F4 antibody binds to an epitope expressed by the p-pleated
sheet configuration of amyloids, just as its murine counterpart does, but
surprisingly, as shown in Example 6 below, the chimeric antibody binds to AL
amyloid fibrils with higher affinity than the 11-1F4 mouse antibody from which
it
was derived.
[00104] One can
also clone the genes for the CDR regions to produce a
humanized form of the antibody using known human antibody sequences. Like
the chimeric form of the 11-1F4 antibody, the humanized form may also have a
binding affinity for amyloid fibrils that is higher than that of the murine
counterpart.
[00105] Those
of skill in the art will understand that the disclosed
humanized and chimeric antibodies may utilize all different types of human
constant regions and/or framework regions. For
example, the disclosed
humanized and chimeric antibodies may comprise the constant regions and/or
framework regions of a human IgG (including IgG1, IgG2, IgG3, and IgG4), IgA,
IgE, IgH, or IgM. In preferred embodiments, the disclosed humanized or
chimeric 11-1F4 antibody comprises a human IgG1 constant region.
[00106] In some
embodiments, the disclosed antibodies may comprise one
or more substitutions, insertions, or deletions, so long as the antibody
maintains
the ability to bind to amyloid fibrils (e.g., kappa and/or lambda light chain
fibrils).
For example, in some embodiments, a chimeric 11-1F4 antibody of the present
disclose may comprise heavy and light chains with about 85%, about 86%, about
87%, about 88%, about 89%, about 90%, about 91%, about 92%, about 93%,
about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about
23

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
100% identity compared to the corresponding heavy and light chain sequences
disclosed herein, so long as the antibody maintains the ability to bind to
amyloid
fibrils. In some embodiments, a humanized 11-1F4 antibody of the present
disclose may comprise CDRs that have about 85%, about 86%, about 87%,
about 88%, about 89%, about 90%, about 91%, about 92%, about 93%, about
94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%
identity compared to the corresponding CDR sequences disclosed herein, so
long as the antibody maintains the ability to bind to amyloid fibrils.
[00107] In some embodiments, the disclosed humanized or chimeric
antibody binds to amyloid fibrils with a higher affinity than its murine
equivalent in
vitro and/or in vivo as determined by, for example, the direct binding ELISA
assay described in Example 6. Without being bound by theory, it is thought
that
the disclosed humanized and chimeric 11-1F4 antibodies can bind and neutralize

toxic circulating amyloid proteins that have not yet formed deposits or
fibrils, and
the disclosed humanized and chimeric antibodies can dissolve amyloid deposits.

Indeed, chimeric 11-1F4 antibodies were demonstrated to bind to fibrils and to

dissolve human amyloidomas in mice. This is of note because it is believed
that
the precursor light chain protein is toxic to cardiomyocytes, and therefore a
treatment approach that can target the circulating toxic amyloid precursor
proteins as well the aggregated amyloid fibrils deposited within organs could
improve cardiovascular outcomes in patients with AL Amyloidosis that has
myocardial involvement, even if the patient's disease is not hematologically
controlled (i.e., the patient has detectable levels of toxic amyloid precursor

proteins in the patient's blood or serum or the difference between involved
and
uninvolved free light chains of >40 mg/L).
Abbreviations
[00108] Dulbecco's Modified Eagles Medium (DMEM), Fetal Bovine
Serum (FBS), ribonucleic acid (RNA); messenger RNA (mRNA);
deoxyribonucleic acid (DNA); copy DNA (cDNA); polymerase chain reaction
(PCR); minute (min); second (sec); Tris-borate buffer (TBE).
24

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[00109] Amino
acids are represented by the IUPAC abbreviations, as
follows: Alanine (Ala), Arginine (Arg), Asparagine (Asn), Aspartic acid (Asp),

Cysteine (Cys), Glutamine (Gin), Glutamic acid (Glu), Glycine (Gly), Histidine

(His), Isoleucine (Ile), Leucine (Leu), Lysine (Lys), Methionine (Met),
Phenylalanine (Phe), Proline (Pro), Serine (Ser), Threonine (Thr), Tryptophan
(Trp), Tyrosine (Tyr), Valine (Val). Similarly for nucleotides: Adenine (A),
Cytosine (C), Guanine (G), Thymine (T), Uracil (U),Adenine or Guanine (R),
Cytosine or Thymine (Y), Guanine or Cytosine (S), Adenine or Thymine (W),
Guanine or Thymine (K), Adenine or Cytosine (M), Cytosine or Guanine or
Thymine (B), Adenine or Guanine or Thymine (D), Adenine or Cytosine or
Thymine (H), Adenine or Cytosine or Guanine (V), and any base (N).
Humanized or Chimeric Antibodies
[00110] To
produce the chimeric antibodies of the invention, the murine
11-1F4 monoclonal antibody heavy and kappa light chain variable region
genes described in United States patent 8,105,594 were PCR modified to
facilitate the expression of the chimeric 11-1F4 antibody in mammalian cells.
A detailed sequence analysis of the modified variable region genes was
performed. The
modified variable region genes were cloned into the
appropriate mammalian expression vectors, creating the constructs 11-
1 F4VHpG1D200 and 11-1F4VK.pKN100. A single supervector construct,
pG1KD200-11-1F4, was made from the 11-1F4VHpG1D200 and 11-
IF4VK.pKN100 constructs by EcoRI restriction enzyme digest and ligation.
Finally, the chimeric 11-1F4 antibody was transiently expressed in COS cells
by both cotransfection and single supervector transfection. While COS cells
were chosen for the co-transfection or transfection as a matter of
convenience,
those of skill in the art would recognize that other mammalian cell lines
could
be used. The characterization of the binding capacity of the chimeric 11-1F4
antibody for amyloid fibrils was determined by direct binding ELISA.
Unexpectedly and beneficially, the chimeric 11-1F4 antibody bound to amyloid
fibrils with higher affinity than the murine 11-1F4 antibody.

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[00111] Typically, an antibody consists of four polypeptides: two
identical
copies of a heavy (H) chain polypeptide and two copies of a light (L) chain
polypeptide. Typically, each heavy chain contains one N-terminal variable (VH)

region and three C-terminal constant (CH1, CH2 and CH3) regions, and each
light chain contains one N-terminal variable (VL or VK) region and one C-
terminal constant (CL) region. The variable regions of each pair of light and
heavy chains form the antigen binding site of an antibody.
[00112] An antibody useful in the compositions and methods of the
invention may be a chimeric mouse-human monoclonal antibody comprising
the VK region of SEQ ID NO: 47 and the VH region of SEQ ID NO: 48 or a
humanized monoclonal antibody comprising CDR sequences of SEQ ID NOs:
49-54. These antibodies bind to an epitope expressed by the p-pleated sheet
configuration of amyloid fibrils. Moreover, surprisingly the antibodies bind
to
this epitope with higher affinity than the 11-1F4 mouse antibody from which
they were derived, which comprises the VK region of SEQ ID NO: 36 and the
VH region of SEQ ID NO: 35. The invention includes methods of treating an
amyloid deposition disease in a human patient in need of such treatment
which comprises administering to the patient a therapeutically effective dose
of
one of the above antibodies in a pharmaceutically-acceptable carrier. The
amount of antibody administered should be effective, for example, to reduce
the amount of amyloid fibrils deposited in the tissues of the patient. The
antibody composition may be administered by any conventional route of
administration, but parenteral administration (such as intravenous) is
preferred. Pharmaceutically-acceptable carriers are well-known in the art and
a suitable one can be selected by one of skill in the medical field. The
amyloid
deposition disease is preferably primary (AL) amyloidosis.
[00113] A chimeric antibody useful for the compositions and methods of
the described and claimed herein (and a method of making the chimeric
antibody) is disclosed in co-owned Patent Cooperation Treaty application
__________ (docket 8441-0004W0 with priority to United States patent
application 62/526,835, filed June 29, 2017), filed on even date herewith and
26

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
incorporated herein in its entirety. Materials
useful to make the subject
antibody include vector constructs selected from the group consisting of 11-
1F4VK.pKN100 and 11-F4VH.pG1D200, shown in Figures 5 and 6,
respectively, and the superconstruct pG.1KD20011-1F4 made from the two
above vector constructs. Other useful materials include the modified murine
11-1F4 antibody VK region gene (SEQ ID NO: 42) and the modified 11-1F4
antibody VH region gene (SEQ ID NO: 45), as well as the respective primers
SEQ ID NO: 41, 43, 44, and 46. The subject antibody may be made by co-
transfection of the vector constructs 11-1F4VK.pKN100 and 11-
F4VH.pG1D200 or the superconstruct pG.1KD20011-1F4 in a suitable
mammalian host cell, such as COS (Chinese hamster ovary) cells.
[00114] Methods
of making, testing, and using the humanized or chimeric
11-1F4 antibody are discussed in further detail in the Examples section below.
Pharmaceutical Formulations
[00115]
Pharmaceutical compositions suitable for use in the methods
described herein can include the disclosed humanized or chimeric 11-1F4
antibodies, humanized antibodies, or antigen-binding antibody fragments and
a pharmaceutically acceptable carrier or diluent.
[00116] The
composition may be formulated for intravenous,
subcutaneous, intraperitoneal, intramuscular, oral, nasal, pulmonary, ocular,
vaginal, or rectal administration. In some embodiments, the antibodies are
formulated for intravenous, subcutaneous, intraperitoneal, or intramuscular
administration, such as in a solution, suspension, emulsion, liposome
formulation, etc. The pharmaceutical composition can be formulated to be an
immediate-release composition, sustained-release composition, delayed-
release composition, etc., using techniques known in the art.
[00117]
Pharmacologically acceptable carriers for various dosage forms
are known in the art. For example, excipients, lubricants, binders, and
disintegrants for solid preparations are known; solvents, solubilizing agents,

suspending agents, isotonicity agents, buffers, and soothing agents for liquid
27

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
preparations are known. In some
embodiments, the pharmaceutical
compositions include one or more additional components, such as one or
more preservatives, antioxidants, stabilizing agents and the like.
[00118]
Additionally, the disclosed pharmaceutical compositions can be
formulated as a solution, microemulsion, liposome, or other ordered structure
suitable to high drug concentration. The carrier can be a solvent or
dispersion
medium containing, for example, water, ethanol, polyol (for example, glycerol,

propylene glycol, and liquid polyethylene glycol, and the like), and suitable
mixtures thereof. The proper fluidity can be maintained, for example, by the
use of a coating such as lecithin, by the maintenance of the required particle

size in the case of dispersion and by the use of surfactants. In some
embodiment, it will be preferable to include isotonic agents, for example,
sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the
composition. Prolonged absorption of the injectable compositions can be
brought about by including in the composition an agent that delays absorption,

for example, monostearate salts and gelatin.
[00119] Sterile
injectable solutions can be prepared by incorporating the
active compound in the required amount in an appropriate solvent with one or
a combination of ingredients enumerated above, as required, followed by
sterilization microfiltration. Generally, dispersions are prepared by
incorporating the active compound into a sterile vehicle that contains a basic

dispersion medium and the required other ingredients from those enumerated
above. In the case of sterile powders for the preparation of sterile
injectable
solutions, the preferred methods of preparation are vacuum drying and freeze-
drying (Iyophilization) that yield a powder of the active ingredient plus any
additional desired ingredient from a previously sterile-filtered solution
thereof.
[00120]
Pharmaceutical compositions of the disclosure can be
administered in combination with other therapeutics that are part of the
current
standard of care for amyloidosis and amyloid diseases. Alternatively, the
disclosed pharmaceutical compositions may be administered to a patient that
28

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
has previously received conventional treatment for amyloidosis and amyloid
diseases, but who has not responded to conventional treatment (i.e., the
disease is refractory or continues to progress).
Methods of Treatment
[00121] In the
present invention, at least one chimeric antibody,
humanized antibody, or antigen-binding antibody fragment thereof is
administered to a patient (e.g., a human patient) suffering from amyloidosis
to
promote the degradation and removal of at least some of the amyloid fibrils
which have become deposited in the organs of the patient and/or which are
circulating in the patient's bloodstream. In some embodiments, therapeutically

effective amount of the antibody is administered together with a
pharmaceutically acceptable carrier. Suitable pharmaceutically acceptable
carriers are well-known in the art, as discussed infra. A typical
route of
administration is parenterally (e.g., intravenously, subcutaneously, or
intramuscularly), as is well understood by those skilled in the medical arts.
Other routes of administration are, of course, possible. Administration may be

by single or multiple doses. The amount of antibody administered and the
frequency of dosing may be optimized by the physician for the particular
patient.
[00122]
Amyloidosis can affect different organs in different people, and
there are different types of amyloid. Amyloidosis frequently affects the
heart,
kidneys, liver, spleen, nervous system and digestive tract. Severe amyloidosis

can lead to life-threatening organ failure.
[00123] Signs and
symptoms of amyloidosis may include, but are not
limited to: swelling of the ankles and legs; severe fatigue and weakness;
shortness of breath; numbness, tingling or pain in the hands or feet,
especially
pain in the wrist (carpal tunnel syndrome); diarrhea, possibly with blood, or
constipation; unintentional, significant weight loss; an enlarged tongue; skin

changes, such as thickening or easy bruising, and purplish patches around the
eyes; an irregular heartbeat; or difficulty swallowing.
29

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[00124] In general, amyloidosis is caused by the buildup of an abnormal
protein called amyloid. Amyloid is produced in the bone marrow and can be
deposited in any tissue or organ. The specific cause of the condition depends
on the type of amyloidosis.
[00125] There are several types of amyloidosis or amyloid diseases,
including AL amyloidosis, AA amyloidosis, and hereditary amyloidosis.
[00126] AL amyloidosis (immunoglobulin light chain amyloidosis) is the
most common type and can affect the heart, kidneys, skin, nerves and liver.
Previously known as primary amyloidosis, AL amyloidosis occurs when the
bone marrow produces abnormal antibodies that cannot be broken down. The
antibodies are deposited in various tissues as amyloid plaques, which
interfere
with normal function of the tissue or organ.
[00127] AA amyloidosis generally affects the kidneys but occasionally
also affects the digestive tract, liver or heart. It was previously known as
secondary amyloidosis. It often occurs along with chronic infectious or
inflammatory diseases, such as rheumatoid arthritis or inflammatory bowel
disease.
[00128] Hereditary amyloidosis (familial amyloidosis) is an inherited
disorder that usually often affects the liver, nerves, heart, and/or kidneys.
Many different types of gene abnormalities present at birth are associated
with
an increased risk of amyloid disease or hereditary amyloidosis. The type and
location of an amyloid gene abnormality can affect the risk of certain
complications, the age at which symptoms first appear, and the way the
disease progresses over time.
[00129] When an amyloid disease affects the heart, it can cause
numerous types of complications. Amyloid deposits or plaques reduce the
heart's ability to fill with blood between heartbeats. Less blood is pumped
with
each beat, and this may lead to shortness of breath. Amyloid deposits or
plaques in or around the heart may also cause irregular heartbeats and
congestive heart failure, among other organ dysfunctions

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[00130] When an
amyloid disease affects the kidneys, it will often harm
the kidneys' filtration ability, allowing protein to leak from the blood into
the
urine (i.e., proteinuria). Moreover,
the kidneys' ability to remove waste
products from your body is lowered, which may eventually lead to kidney
failure.
[00131] Provided
herein are methods of treating an amyloid deposition
diseases, such as primary (AL) amyloidosis, in a patient (e.g., a human
patient) in need of such treatment which comprises administering to the
patient
a humanized or chimeric 11-1F4 antibody together with a pharmaceutically
acceptable carrier, in an amount effective to treat the amyloid deposition
disease.
[00132] In some
embodiments, the amyloid deposition disease (e.g.,
primary amyloidosis) comprises involvement of at least one organ or tissue
selected from the group consisting of heart, kidneys, liver, lung,
gastrointestinal tract, nervous system, muscular skeletal system, soft tissue,

and skin.
[00133] In
embodiments when the disease involves amyloid deposits or
plaques in the patient's heart, treatment with the disclosed humanized or
chimeric 11-1F4 antibody may decrease the levels of the patient's N-terminal
pro b-type natriuretic peptide (NT-proBNP) by at least about 30% compared to
baseline levels taken prior to administration of the antibody. In some
embodiments, the decrease in NT-proBNP may be at least about 40%, at least
about 50%, at least about 60% or more compared to baseline levels taken
prior to administration of the antibody. In some embodiments, treatment with
the disclosed human or chimeric 11-1F4 antibody may result in the patient's
NT-proBNP level decreasing to less than about 9100 ng/L following
administration of the antibody. In some embodiments, the patient's NT-
proBNP level may decrease to less than about 8000, 7000, 6000, 5000, or
4000 ng/L following administration of the antibody. In some embodiments, the
patient may initially be classified as New York Heart Association (NYHA)
31

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
Functional Classification class II or III prior to administration of the
antibody,
but after treatment with the disclosed chimeric 11-1F4 antibody the patient
may be classified as class I on the NYHA classification scale.
[00134]
Provided herein are methods of improving myocardial function in
patient suffering from amyloidosis with cardiac involvement, as well as
methods
for treating specific sub-populations of patients, such as those with ALA that
has
cardiac involvement (i.e., NT-proBNP is greater than 650 pg/mL) and that is
not
hematologically controlled.
[00135] When an
amyloid disease affects the heart, it can cause numerous
types of complications and this cardiac involvement portends poor prognosis.
Amyloid deposits or plaques reduce the heart's ability to fill with blood
between
heartbeats. Less blood is pumped with each beat, and this may lead to
shortness
of breath, among other serious complicating factors. Amyloid deposits or
plaques
in or around the heart may also cause irregular heartbeats and congestive
heart
failure, among other organ dysfunctions.
[00136]
Provided herein are methods of improving myocardial function in a
patient diagnosed with amyloid light chain amyloidosis (ALA) with cardiac
involvement comprising administering to a patient diagnosed with ALA with
cardiac involvement a therapeutically effective amount of a humanized or
chimeric 11-1F4 antibody or an antigen-binding fragment thereof. The antibody
or an antigen-binding fragment thereof may bind to amyloid fibrils with higher

affinity than murine 11-1F4 antibody, as determined by direct binding ELISA.
Further, the improvements in myocardial function may be apparent within about
three weeks after administering the antibody or antigen-binding fragment
thereof.
For example, improvement in various measures of myocardial function may be
seen within about 1, about 2, about 3, about 4, about 5, about 6, about 7,
about
8, about 9, about 10, about 11, about 12, about 13, about 14, or about 15
weeks
of the initiation of treatment.
[00137] The
myocardial function of patients having cardiac involvement can
be determined by measuring N-terminal pro b-type natriuretic peptide (NT-
32

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
proBNP) levels. Tissue damage caused by amyloid deposits in the heart of ALA
patients increases NT-proBNP levels in the patients. In some embodiments, the
patients diagnosed with ALA having cardiac involvement exhibits a pre-
treatment
NT-proBNP level greater than 650 pg/mL.
[00138]
Myocardial function and improvement therein can be measured by
using echocardiography to measure global longitudinal strain (GLS) as
described
in Smiseth et al. ¨ Eur Heart J, 37:1196. Echocardiography uses ultrasound to
measure the average deformation within segments of the myocardium, and GLS
is the average of these segments as a measure of global left ventricular
function.
Amyloid deposits can result in thickened left and right ventricular walls and
in a
non-dilated ventricle that is stiff and poorly compliant, resulting in
"strain" on the
heart and vasculature. In echocardiography parlance, the term "strain" is used
to
describe deformation in the myocardium, which may include but is not limited
to
local shortening, thickening, and/or lengthening of the myocardium. Strain can
be
used as a measure of ventricular function. A person having ordinary skill in
the
art will know how to use echocardiography to determine GLS and will understand

that it may be calculated in various ways. For example, the Lagrangian formula

(EL = (L-Lo)/Lo = UL , where Lo is baseline length and L is the resulting
length),
defines strain in relation to the original length as a dimensionless measure,
in
which shortening will be negative, and lengthening will be positive. It is
usually
expressed in percent. An alternative definition, Eulerian strain, defines the
strain
in relation to the instantaneous length: EE = AL/L. For a change over time,
the
Lagrangian strain will be: EL = >AL/Lo, and Eulerian Strain EE = > (UL). The
term was first used by Mirsky and Parmley in describing regional differences
in
deformation between normal and ischemic myocardium.
[00139] Hence,
in some embodiments, the patients of the presently
disclosed method exhibits an improvement in global longitudinal strain (GLS)
compared with pretreatment GLS level. For instance, in a study of 19 patients
treated with the disclosed chimeric antibody, 10 of these patients had cardiac

involvement per screening NT-proBNP levels, and 8 patients were cardiac
evaluable per baseline NT-proBNP, wherein cardiac involvement is defined by
33

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
having NT-proBNP level greater than 650 pg/mL. In some embodiments, a
patient with ALA with cardiac involvement may have a baseline NT-proBNP of at
least 650 pg/ml, while in some embodiments, a patient with ALA with cardiac
involvement may have a baseline NT-proBNP of at least 700, 750, 800, 850, 900,

950, 1000, 1050, 1100, 1150, 1200, 1250, 1300, 1350, 1400, 1450, 1500, 1550,
1600, 1650, 1700, 1750, 1800, 1850, 1900, 1950, 2000, 2050, 2100, 2150, 2200,
2250, or 2300 or more pg/m I.
[00140] As
disclosed in Example 9 below, 9 out of the 10 patients having
cardiac involvement exhibited improvement in myocardial function upon
exposure to the disclosed chimeric antibody as shown in Figure 15. Hence, in
some embodiments, the patient treated with a therapeutically effective dose of

the chimeric antibody exhibits an improvement in global longitudinal strain
(GLS)
compared with pretreatment GLS level. Concomitant with reduced GLS, the
patients treated with the disclosed antibodies may also exhibit reduced NT-
proBNP levels.
[00141] In some
embodiments, the improvement in GLS may occur within
about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about
9,
about 10, about 11, about 12, about 13, about 14, or about 15 weeks of the
initiation of treatment. In some embodiments, the improvement in GLS may be
represented by a 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17,
18, 19, 20,
21, 22, 23, 24, 25, or more percent reduction in GLS compared to baseline as
calculated by the Lagrangian formula. A reduction in GLS level compared to
baseline of about 2% percent or more is considered clinically relevant.
[00142] In some
embodiments, the disclosed treatments with the
humanized or chimeric 11-1F4 antibody or an antigen-binding fragment thereof
may decrease the levels of the patient's N-terminal pro b-type natriuretic
peptide
(NT-proBNP) by at least about 30% compared to baseline levels taken prior to
administration of the antibody. In some embodiments, the decrease in NT-
proBNP may be at least about 40%, at least about 50%, at least about 60% or
more compared to baseline levels taken prior to administration of the
antibody.
34

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
In some embodiments, treatment with the disclosed chimeric 11-1F4 antibody
may result in the patient's NT-proBNP level decreasing to less than about 9100
ng/L following administration of the antibody. In some
embodiments, the
patient's NT-proBNP level may decrease to less than about 8000, 7000, 6000,
5000, or 4000 ng/L following administration of the antibody. In some
embodiments, the patient may initially be classified as New York Heart
Association (NYHA) Functional Classification class II or III prior to
administration
of the antibody, but after treatment with the disclosed chimeric 11-1F4
antibody
the patient may be classified as class I on the NYHA classification scale.
[00143] In some
embodiments, the disclosed methods comprise treating a
patient suffering from relapse or refractory ALA. In some embodiments, the
patient may have kappa ALA. In some embodiments, the patient may have
lambda ALA.
[00144]
Immunoglobulins are composed of four protein chains: two light
chains, either kappa (K) or lambda (A) light chains, and two heavy chains, of
which there are several types. In AL amyloidosis, either kappa light chains or

lambda light chain may be misfolded and form amyloid fibrils or plaques.
Hence,
in some patients, both kappa and lambda fragments may be misfolded.
Subgroup analysis showed that patients with both lambda cardiac involvement
and kappa cardiac involvement showed improvement by having reduced GLS
compared to pre-treatment levels as shown in Example 8. Hence, in some
embodiments the patient is further characterized as having light chain lambda
amyloid cardiac involvement. In some embodiments, the patient is further
characterized as having light chain kappa amyloid cardiac involvement.
[00145] In some
embodiments, the disclosed methods of treatment may
further comprise administering a chemotherapeutic drug that may be intended to

kill the dysfunctional cells that are creating the toxic precursor protein. In
some
cases, such therapy may be successful, thereby resulting in a decrease in
dysfunctional cells and a concomitant decrease in the amount of circulating
toxic
amyloid precursor proteins in the patient's blood. However, in some cases,

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
chemotherapy may be ineffective in decreasing the number of dysfunctional
cells
and/or the ability of these cells to produce the toxic amyloid precursor
proteins.
Those patients that continue to have detectable levels of circulating toxic
amyloid
precursor proteins in their blood are said to have ALA that is not
hematologically
controlled.
[00146] The
disclosed methods of treatment may be particularly beneficial
for patients with disease that is not hematologically controlled (i.e., in
neither
complete remission nor very good partial remission) because the presently
disclosed humanized and chimeric 11-1F4 antibodies are believed to be able to
bind to and neutralize toxic amyloid precursor proteins in circulation, even
before
the proteins aggregate to form an amyloid deposit. Complete remission is
defined as negative serum and urine immunofixation and normal ratio in a free-
light-chain (FLC) assay, while very good partial remission is defined as
having a
difference between involved and uninvolved free light chains of <40 mg/L.
Monitoring improvement
[00147]
Echocardiography is non-invasive and can be used to monitor
improvement in myocardial function in a patient diagnosed with light chain
amyloidosis (ALA) having a cardiac involvement comprising observing an
improvement in myocardial function in a patient diagnosed with ALA having a
cardiac involvement within about three weeks after administration to said
patient
of a therapeutically effective amount of a humanized or chimeric 11-1F4
antibody
(c11-1F4 Ab) or an antigen-binding fragment thereof, said humanized or
chimeric
11-1F4 Ab or an antigen-binding fragment thereof having a binding affinity to
amyloid fibrils, which is higher than that of murine 11-1F4 antibody, as
determined by direct binding ELISA. Hence, the improved myocardial function
can be observed in about three weeks after administration of the humanized or
chimeric 11-1F4 antibody as shown in Example 8. In some embodiments, the
improvement in myocardial function persists for a period extending at least
three
months after administration of the humanized or chimeric 11-1F4 Ab or an
antigen-binding fragment thereof.
36

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[00148] In
embodiments when the disease involves amyloid deposits or
plaques in the patient's kidneys, treatment with the disclosed humanized or
chimeric 11-1F4 antibody may decrease the level of protein in the patient's
urine
(i.e., proteinuria) by at least about 30% compared to baseline levels
determined
prior to administration of the antibody. In some embodiments, the decrease in
protein in the patient's urine may be at least about 40%, at least about 50%,
at
least about 60% or more compared to baseline levels determined prior to
administration of the antibody. In some embodiments, the patient's urine
protein
output may decrease to less than about 7000, less than about 6000, less than
about 5000, less than about 4000, or less than about 3000 mg/24 hours
following
administration of the antibody.
Therapeutically effective doses and dosing regimens
[00149] In some
embodiment, the therapeutically effective dose of the
antibody may be administered no more than once, twice, three, or four times
within a three month period. In some embodiments, the reduction in NT-proBNP
or improvement in GLS is sustained in the patient for at least about three
months
after the administration of a humanized or chimeric 11-1F4 antibody.
[00150]
Therapeutically effective doses and dosing regimens of the
foregoing methods may vary, as would be readily understood by those of skill
in
the art. Dosage regimens may be adjusted to provide the optimum desired
response (e.g., a therapeutic response clearance of amyloid plaques or
reduction
in the amount of deposited amyloid fibrils). For example, in some embodiments,

a single dose of the antibody may be administered, while in some embodiments,
several divided doses may be administered over time, or the dose may be
proportionally reduced or increased in subsequent dosing as indicated by the
situation. For example, in some embodiments the disclosed antibodies may be
administered once or twice weekly by subcutaneous, intravenous, or
intramuscular injection. In some embodiments, the disclosed antibodies or
antigen-binding fragments thereof may be administered once or twice monthly by

subcutaneous, intravenous, or intramuscular injection. In some embodiments,
37

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
the disclosed antibodies or antigen-binding fragments thereof may be
administered once or twice annually by subcutaneous, intravenous, or
intramuscular injection. In some embodiments, the disclosed antibodies or
antigen-binding fragments thereof may be administered once every week, once
every other week, once every three weeks, once every four weeks, once every
month, once every other month, once every three months, once every four
months, once every five months, once every six months, once every seven
months, once every eight months, once every nine months, once every ten
months, once every eleven months, twice a year, or once a year, as the
situation
or condition of the patient may indicate.
[00151]
Moreover, the data disclosed herein indicates that patient response
to treatment with humanized or chimeric 11-1F4 antibody is not only sustained,

but rapid as well. In some embodiments, a patient may experience a therapeutic

response (i.e., a decrease in the size of amyloid deposits or plaques, a
decrease
in the rate of plaque formation, or improved organ function) in one week or
less,
two weeks or less, three weeks or less, four weeks or less, five weeks or
less, six
weeks or less, seven weeks or less, eight weeks or less, nine weeks or less,
ten
weeks or less, eleven weeks or less, twelve weeks or less, or any time frame
in
between. For example, depending on the dose and dosing regimen, a patient
may experience a therapeutic response in about a week or about 4.5 weeks.
[00152] The
therapeutically effective dose of antibody administered to the
patient (whether administered in a single does or multiple doses) should be
sufficient to reduce the amount of deposited amyloid fibrils in the patient.
Such
therapeutically effective amount may be determined by evaluating the
symptomatic changes in the patient or by evaluating the change in the amount
of
deposited amyloid fibrils (e.g., by radioimmune detection of deposited amyloid

deposits using 1241 tagged antibody). Thus, a labeled antibody of the
disclosure
may be used to detect the presence of amyloid deposition disease in a patient
suspected of having the disease as well as to determine the effectiveness of
treatment.
38

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[00153]
Exemplary doses can vary according to the size and health of the
individual being treated, as well as the condition being treated. In some
embodiments, a therapeutically effective amount of a disclosed humanized or
chimeric 11-1F4 antibody may be about 500 mg/m2 or less; however, in some
situations the dose may be higher. In some embodiments, a therapeutically
effective dose may be 10-1000 mg/m2, 25-900 mg/m2, 50-800 mg/m2, 75-700
mg/m2, 100-600 mg/m2, or any value in between. For instance, in some
embodiments, the therapeutically effective amount may be about 1000, about
975, about 950, about 925, about 900, about 875, about 850, about 825, about
800, about 775, about 750, about 725, about 700, about 675, about 650, about
625,about 600, about 575, about 550, about 525,about 500, about 475, about
450, about 425, about 400, about 375, about 350, about 325, about 300, about
275, about 250, about 225, about 200, about 175, about 150, about 125, about
100 or less mg/m2.
[00154]
Similarly, in some embodiments, the effective amount of a
humanized or chimeric 11-1F4 antibody is about 2,200 mg; however, in some
situations the dose may be higher or lower. In some embodiments, a
therapeutically effective amount may be between 50 and 5000 mg, between 60
about 4500 mg, between 70 and 4000 mg, between 80 and 3500 mg, between
90 and 3000 mg, between 100 and 2500 mg, between 150 and 2000 mg,
between 200 and 1500 mg, between 250 and 1000 mg, or any dose in between.
For instance, in some embodiments, the therapeutically effective amount may be

about 50 about 60, about 70 , about 80, about 90, about 100, about 150, about
200, about 250, about 300, about 350, about 400, about 450, about 500, about
550, about 600, about 650, about 700, about 750, about 800, about 850, about
900, about 950, about 1000, about 1100, about 1200, about 1300, about 1400,
about 1500, about 1600, about 1700, about 1800, about 1900, about 2000, about
2100, about 2200, about 2300, about 2400, about 2500, about 2600, about 2700,
about 2800, about 2900, about 3000, about 3100, about 3200, about 3300, about
3400, about 3500, about 3600, about 3700, about 3800, about 3900, about 4000,
39

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
about 4100, about 4200, about 4300, about 4400, about 4500, about 4600, about
4700, about 4800, about 4900, about 5000 or more mg.
[00155]
Similarly, in some embodiments, the effective amount of a
humanized or chimeric 11-1F4 antibody is about 25 mg/kg; however, in some
embodiments, the concentration may be higher or lower. In some embodiments,
the effective amount may be about 1-50 mg/kg, about 5-40 mg/kg, about 10-30
mg/kg, or about 15-25 mg/kg or any value in between. For instance, in some
embodiments, the effective amount may be 1, 2, 3, 4, 5,6, 7, 8, 9, 10, 11, 12,
13,
14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 26, 28, 29, 30, 31, 32,
33, 34,
35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 or more
mg/kg.
[00156] The
disclosed methods of treatment may also be combined with
other known methods of treatment as the situation may require. The current
standard of care for AL amyloidosis, for example, generally involves
autologous
blood stem cell transplantation (ASCT) or an autologous bone marrow
transplant.
Many of the same chemotherapy medications that treat multiple myeloma are
used in AL amyloidosis to stop the growth of abnormal or dysfunctional cells
that
produce amyloid/toxic amyloid precursor proteins. Thus, in some embodiments,
the disclosed humanized or chimeric 11-1F4 antibodies may be administered
before, after, or concurrently with other known treatments. In some
embodiments, the disclosed humanized or chimeric 11-1F4 antibodies may be
administered only after other treatment options have failed or the disease has

continued to progress. In other words, in some embodiments, the disclosed
antibodies are used to treat refractory amyloid disease, such as refractory AL

amyloidosis.
[00157] In embodiments when the disease involves amyloid deposits or
plaques in the patient's kidneys, treatment with the disclosed chimeric 11-1F4

antibody may decrease the level of protein in the patient's urine (i.e.,
proteinuria) by at least about 30% compared to baseline levels determined
prior to administration of the antibody. In some embodiments, the decrease in
protein in the patient's urine may be at least about 40%, at least about 50%,
at

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
least about 60% or more compared to baseline levels determined prior to
administration of the antibody. In some embodiments, the patient's urine
protein output may decrease to less than about 7000, less than about 6000,
less than about 5000, less than about 4000, or less than about 3000 mg/24
hours following administration of the antibody.
[00158] Also provided herein are methods of treating a patient with an
amyloid deposition disease (e.g., AL amyloidosis) comprising administering a
therapeutically effective amount of a humanized or chimeric 11-1F4 antibody
to a patient in need thereof less frequently than once per month. In some
embodiments, the AL amyloidosis may comprise aggregates of lambda light
chain fibrils, in which case the presence of aggregates of lambda light chain
fibrils is significantly reduced following administration of the antibody.
[00159] In some embodiment, the therapeutically effective dose of the
antibody may be administered once every two months, once every three
months, once every four months, once every five months, once every six
months, or biannually, and more specific dosing regimens are discussed
below.
[00160] Also provided herein are methods of treating a patient with
primary light chain (AL) amyloidosis involving the heart comprising
administering to said patient a dose of a humanized chimeric 11-1F4 antibody,
said dose being effective to cause at least a 30% reduction in N-terminal pro
b-type natriuretic peptide (NT-proBNP) level following administration of the
humanized or chimeric 11-1F4 antibody as compared to pre-treatment level.
Also included are methods of treating a patient with primary light chain (AL)
amyloidosis involving the kidneys comprising administering to said patient a
dose of a humanized or chimeric 11-1F4 antibody, said dose effective to cause
at least a 40% reduction in proteinuria following administration of the
humanized or chimeric 11-1F4 antibody compared to pre-treatment levels.
41

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[00161] In some
embodiments, the reduction in NT-proBNP and/or
proteinuria is sustained in the patient for at least about six months after
the
administration of the humanized or chimeric 11-1F4 antibody.
[00162] As
described above, immunoglobulins are composed of four
protein chains: two light chains, either kappa (K) or lambda (A) light chains,
and
two heavy chains, of which there are several types. In AL amyloidosis, either
kappa light chains or lambda light chain may be misfolded and form amyloid
fibrils or plaques. In some patients, both kappa and lambda fragments may be
misfolded. Thus, provided herein are methods of decreasing the amount of
kappa and/or lambda light chain fibril aggregate deposits in a patient in need

thereof comprising, administering to a patient with primary amyloidosis
comprising kappa or lambda light chain fibril aggregate deposits a dose of an
antibody comprising: (i) a VK region comprising SEQ ID NO: 47, a VH region
comprising SEQ ID NO: 48 or (ii) CDR sequences comprising SEQ ID NOs:
49-54, and a human IgG1 constant region; wherein the does is effective to
decrease the amount of kappa or lambda light chain fibril aggregate deposits
in the patient.
[00163] In some
embodiments, the primary amyloidosis consists of
lambda light chain fibril deposits or plaques, while in some embodiments, the
primary amyloidosis consists of kappa light chain fibril aggregate deposits,
and
in some embodiments, the primary amyloidosis consists of kappa and lambda
light chain fibril aggregate deposits.
[00164] It was
surprisingly discovered herein that chimeric 11-1F4 is
unexpectedly efficacious in clearing lambda light chain fibrils. Indeed,
preclinical experiments such as the mouse study provided in Example 7
suggested that lambda fibrils were resistant to clearance and would
requirement multiple recurrent treatments in order for the deposits to clear.
However, as shown in Example 8, when the chimeric 11-1F4 antibody was
administered to humans, the treatment resulted in a decrease in lambda chain
amyloid deposits after only a single dose. This result was entirely unexpected
42

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
prior to performing the disclosed methods. It is believed that a humanized 11-
1F4 antibody would exhibit a similar capacity to clear lambda light chain
fibrils.
[00165] In any of the foregoing methods, administration of the disclosed
humanized or chimeric 11-1F4 antibody is expected to decrease organ
dysfunction. Additionally, in any of the foregoing methods, the constant
region
of the antibody may be a human IgG constant region. More specifically, in
some embodiments, the constant region of the antibody may be a human IgG1
constant region.
[00166] Therapeutically effective doses and dosing regimens of the
foregoing methods may vary, as would be readily understood by those of skill
in the art. Dosage regimens may be adjusted to provide the optimum desired
response (e.g., a therapeutic response clearance of amyloid plaques or
reduction in the amount of deposited amyloid fibrils). For example, in some
embodiments, a single bolus dose of the antibody may be administered, while
in some embodiments, several divided doses may be administered over time,
or the dose may be proportionally reduced or increased in subsequent dosing
as indicated by the situation. For example, in some embodiments the
disclosed antibodies may be administered once or twice weekly by
subcutaneous, intravenous, or intramuscular injection. In some embodiments,
the disclosed antibodies or functional fragments may be administered once or
twice monthly by subcutaneous, intravenous, or intramuscular injection. In
some embodiments, the disclosed antibodies or functional fragments may be
administered once or twice annually by subcutaneous, intravenous, or
intramuscular injection. In some embodiments, the disclosed antibodies may
be administered once every week, once every other week, once every three
weeks, once every four weeks, once every month, once every other month,
once every three months, once every four months, once every five months,
once every six months, once every seven months, once every eight months,
once every nine months, once every ten months, once every eleven months,
twice a year, or once a year, as the situation or condition of the patient may

indicate.
43

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[00167] Moreover, the data disclosed herein indicates that patient
response to treatment with humanized or chimeric 11-1 F4 antibody is not only
sustained, but also rapid as well. In some embodiments, a patient may
experience a therapeutic response (i.e., a decrease in the size of amyloid
deposits or plaques, a decrease in the rate of plaque formation, or improved
organ function) in one week or less, two weeks or less, three weeks or less,
four weeks or less, five weeks or less, six weeks or less, seven weeks or
less,
eight weeks or less, nine weeks or less, ten weeks or less, eleven weeks or
less, twelve weeks or less, or any time frame in between. For example,
depending on the dose and dosing regimen, a patient may experience a
therapeutic response in about a week or about 4.5 weeks.
[00168] The following examples are given to illustrate the present
invention. It should be understood, however, that the invention is not to be
limited to the specific conditions or details described in these examples. All

printed publications referenced herein are specifically incorporated by
reference.
EXAMPLE 1
PCR Cloning and DNA Sequencing of the mouse 11-1F4 Antibody
[00169] The murine 11-1F4 monoclonal antibody heavy and light chain
variable region genes were PCR cloned and a detailed sequence analysis of
all variable region genes isolated (both pseudo and functional) was performed.

Detailed DNA and amino acid sequences of the murine 11-1F4 antibody heavy
and light chain variable region genes were obtained.
Materials
[00170] Media components and all other tissue culture materials were
obtained from Life Technologies (UK). The RNA solution kit was obtained
from Stratagene (USA), while the first strand cDNA synthesis kit was
purchased from Pharmacia (UK). All the constituents and equipment for the
RCR-reaction, including AmpliTaq DNA polymerase, were purchased from
44

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
Perkin Elmer (USA). The TOPO TA Cloning @ kit was obtained from
Invitrogen (USA). Agarose (UltraPureTM) was obtained from Life Technologies
(UK). The ABI PRISM Big DyeTM terminator cycle sequencing ready reaction
kit pre-mixed cycle sequencing kit and the ABI PRISM 310 sequencing
machine were both purchased from PE Applied Biosystems (USA). All other
molecular biological products were obtained from New England Biolabs (USA)
and Promega (USA).
Methods
[00171] The strategy used to PCR clone the murine VH and VK genes
from the hybridoma cell lines producing the murine monoclonal antibody 11 -
1F4 is outlined in Figure 1.
[00172] Two clones (B2C4 and B2D6) of the 5P2/0 hybridoma cell line
producing the a-human light chain monoclonal antibody 11-1F4, were kindly
provided by Alan Solomon, MD (University of Tennessee Medical Center at
Knoxville, TN). The hybridoma cell line is available from the American Type
Culture Collection (ATCC access PTA-105). The cell lines were cultured using
DMEM media supplemented with 20% (v/v) FBS, penicillin/streptomycin and L-
Glutamine. Cells were cultured until a total viable cell count of 108 cells
was
reached.
[00173] The cells were harvested separately from each clone as follows.
The mouse hybridoma cell line was grown in suspension in an appropriate
culture medium and in sufficient quantities to provide a total viable cell
count of
about 108 cells. The culture supernatant was harvested and the hybridoma
cells pelleted in a bench top centrifuge (250 g, 5 min). The cells were gently

re-suspended in 20 ml PBS and a 100 pI aliquot was taken for a viable cell
count. The cells in the aliquot were pelleted once more and 200 pI of PBS and
200 pl of trypan blue were added to the 100 pI of cells and mixed gently. Ten
pI of this mixture was pipetted into a disposable cell-counting slide and the
number of white cells in 9 small squares was counted under a microscope.
Blue cells (i.e., dead cells) were not counted. The count process was

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
repeated, the results averaged, and the average results multiplied by 9 x 105
to obtain a viable cell count for the cells in 20m1 PBS. Once sufficient cells

had been harvested, they were re-suspended in 10 ml of Solution D for RNA
isolation (see below, Stratagene RNA Isolation Kit).
[00174] Total RNA was then isolated separately from the cells of each
clone using a Stratagene RNA isolation kit, according to the manufacturer's
instructions. One ml of 2 M sodium acetate (pH 4.0) was added to the sample
and the contents of the tube were thoroughly mixed by repeatedly inverting the

tube. To the tube was added 10.0 ml of phenol (pH 5.3-5.7) and the contents
again mixed thoroughly by inversion. To the mixture was added 2.0 ml of
chloroform-isoamyl alcohol mixture, the tube was capped and vigorously
shaken for 10 seconds, and the tube was incubated in ice for 15 minutes. The
sample was transferred to a 50-ml thick-walled, round-bottom centrifuge tube
that had been pre-chilled on ice and the tube was spun in a centrifuge at
10,000 x g for 20 minutes at 4 C. Two phases were visible in the tube after
centrifugation. The upper, aqueous phase contained the RNA, while the lower
phenol phase and interphase contained DNA and proteins. The RNA-
containing upper, aqueous phase was transferred to a fresh centrifuge tube
and the lower phenol phase was discarded. An equal volume of isopropanol
was added to the aqueous phase and the contents mixed by inversion,
following which the tube was incubated for 1 hour at -20 C to precipitate the
RNA. The tube was spun in a centrifuge at 10,000 x g for 20 minutes at 4 C.
After centrifugation, the pellet at the bottom of the tube, which contains the

RNA, was removed and the supernatant discarded. The pellet was dissolved
in 3.0 ml of solution D, 3.0 ml of isopropanol was added to the tube and the
contents mixed well. After incubating the tube for 1 hour at -20 C, it was
again
spun in a centrifuge at 10,000 x g for 10 minutes at 4 C and the supernatant
removed from the tube and discarded. (Note: Up to this point. the RNA had
been protected from ribonucleases by the presence of guanidine
isothiocyanate but was now no longer protected.) The pellet was washed with
75% (v/v) ethanol (DEPC-treated water (25%)) and the pellet was dried under
46

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
vacuum for 2-3 minutes. The RNA pellet is re-suspended in 0.5-2 ml of
DEPC-treated water.
[00175] Following the manufacturer's instructions, an Amersham
Pharmacia Biotech first strand cDNA synthesis kit was employed to produce a
single- stranded DNA copy of the 11-1F4 hybridoma mRNA using the Not I-
d(T)18 primer supplied with the kit. One reaction was performed for each of
the
two RNA samples isolated, as follows. The components used were: Bulk first
strand cDNA reaction mix, Cloned FPLCpure TM Murine Reverse Transcriptase,
RNAguardTM, BSA, dATP, dCTP, dGTP, and dTTP, 200 mM DIT aqueous
solution, Not I-d(T)18 primer: 5'
d[AACTGGAAGAATTCGCGGCCGCAGGAA18]-3', and DEPC treated water.
[00176] Approximately 5 pg of total RNA in 20 pI DEPC water was
heated to 65 'C for 10 min and then chilled on ice. The bulk first strand cDNA

reaction mix was pipetted gently to obtain a uniform suspension and the
reaction set up in a 0.5 ml microcentrifuge tube as below. 20 pI denatured
RNA solution, 11 pI Bulk first strand cDNA reaction mix, 1 pI Not I-d(T)18
primer, and 1 ul DTT solution for 33 pl total volume. The reactants were
mixed gently by pipetting and incubated 37 C for 1 hour.
[00177] The murine heavy and kappa light chain variable region genes
(VH genes and VK genes, respectively) were then PCR amplified from the
ssDNA template using the method described by Jones and Bendig
(Bio/Technology 9:88).
[00178] Separate PCR reactions were prepared for each of the
degenerate leader sequence specific primers (MHVI - MHV12 for VH and MKVI
¨ MKV11 for VK) with the appropriate constant region primer (an equimolar mix
of MHCI - MHC3 for VH and MKC for VK). Tables 1 & 2 detail the primers used
to amplify the VH and VK region genes, respectively. In total, 12 heavy chain
reactions and 11 kappa light chain reactions were performed. AmpliTaq
DNA polymerase was used to amplify the template cDNA in all cases, as
follows.
47

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[00179] The
completed cDNA first strand synthesis reaction was heated
at 90 C for 5 minutes to denature the RNA-cDNA duplex and inactivate the
reverse transcriptase and chilled on ice. Eleven GeneAmpTM PCR
reaction tubes were labeled MKV1-11. For each tube a 100 pl reaction
mixture was prepared, each reaction mixture containing 69.3 pl of sterile
water, 10 pl of 10 X PCR buffer II, 6 pl of 25 mM MgCl2, 2 pl each of the 10
mM stock solutions of dNTPs, 2.5 pl of 10 mM MKC primer, 2.5 pl of one of
the 10 mM MKV primers and 1 pl of RNA-cDNA template mix. To each of
the tubes was then added 0.7 pl of AmpliTaq@ DNA polymerase and the
completed reaction mix overlaid with 50 pl of mineral oil.
[00180] A similar
series of reaction mixes was prepared as described
above to PCR-clone the mouse heavy chain variable region gene. However,
this time twelve reaction tubes were labeled and one of the twelve MHV
primers and the appropriate MHC primer were added to each. That is, to
PCR-amplify the variable domain gene of a mouse y1 heavy chain, for
example, the MHC G1 primer was used.
[00181] The
reaction tubes were loaded into a DNA thermal cycler and
cycled (after an initial melt at 94 C for 1.5 min) at 94 C for 1min, 50 C
for I
min and 72 C for 1 min over 25 cycles. The last cycle was followed by a final

extension step at 72 C for 10 min before cooling to 4 C. Except for between
the annealing (50 C) and extension (72 C) steps when an extended ramp
time of 2.5 min was used, a 30 sec ramp time was used between each step of
the cycle. A 10 pl aliquot from each PCR reaction was run on a 1% (w/v)
agarose /1 X TBE buffer gel containing 0.5 pg/ml ethidium bromide to
determine which of the leader primers produced a PCR-product. Positive
PCR-clones were about 420-500 bp in size.
[00182] The above
PCR-amplification process was repeated twice more
and those PCR-reactions that appeared to amplify full-length variable domain
gene were selected. A 6 pl aliquot of each potential PCR-product was directly
cloned into the pCRTm II vector provided by the TA Cloning kit, as described
48

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
in the manufacturers instructions. Aliquots of 10.0% (v/v), 1.0% (v/v) and
0.1%
(v/v) aliquots of the transformed E.coli cells were pipetted onto individual
90
mm diameter LB agar plates containing 50 pg/ml ampicillin, overlaid with 25 pl

of the X-Gal stock solution and 40 pl of IPTG stock solution, and incubated
overnight at 37 C. Positive colonies were identified by PCR-screening.
Table 1 ¨ PCR primers for cloning mouse kappa light chain variable region
genes
Name Sequence (5'->31 SEQ ID
NO:
MICV1 (31mer) ATGAAGATTGCCTGTTAGGCTGTTGGTGCTG 1
MKV2 (30mer) ATGGAGWCAGACACACTCCTGYTATGGGTG 2
MKV3 (30mer) ATGAGTGTGCTCACTCAGGTCCTGGSGTTG 3
MKV4 (33mer) ATGAGGRCCCCTGCTCAGVVTTYTTGGMVVTCTTG 4
MKV5 (30mer) ATGGATTTWCAGGTGCAGATTVVTCAGCTTC 5
MKV6 (29mer) ATGAGGTKCYYTGYTSAYCTYCTCTGRGG 6
MKV7 (32mer) ATGGGCVVTCAAAGATGGAGTCACAKVVYYCWGG 7
MKV8 (30mer) ATGTGGGGAYCTKTTTYCMMTTTTTCAATG 8
MKV9 (25mer) ATGGTRTCCWCASCTCAGTTCCTTG 9
MKV10 (27mer) ATGTATATATGTTTGTTGTCTATTTCT 10
MKV11 (28mer) ATGGAAGCCCCAGCTCAGCTTCTCTTCC 11
MKC (20mer) ACTGGATGGTGGGAAGATGG 12
Table 2¨ PCR primers for cloning mouse heavy chain variable region
genes
Name Sequence (5'->31 SEQ ID
NO:
MHVI (27mer) ATGAAATGCAGCTGGGGCATSTTCTTC 13
MHV2 (26mer) ATGGGATGGAGCTRTATCATSYTCTT 14
MHV3 (27mer) ATGAAGVVTGTGGTTAAACTGGGTTTTT 15
49

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
Name Sequence (5'->31 SEQ ID
NO:
MHV4 (25m er) ATGRACTTTGGGYTCAGCTTGRTTT 16
MHV5 (32mer) ATGGGACTCCAGGCTTCAATTTAGTTTTCCTT 17
MHV6 (29m er) ATGGCTTGTCYTTRGSGCTRCTCTTCTGC 18
MHV7 (27m er) ATGGRATGGAGCKGGRGTCTTTMTCTT 19
MHV8 (23m er) ATGAGAGTGCTGATTCTTTTGTG 20
MHV9 (31m er) ATGGMTTGGGTGTGGAMCTTGCTTATTCCTG 21
M HV10 (28mer) ATGGGCAGACTTACCATTCTCATTCCTG 22
M HV11 (28mer) ATGGATTTTGGGCTGATTTTTTTTATTG 23
MHV12 (27mer) ATGATGGTGTTAAGTCTTCTGTACCTG 24
MHCG1 (21mer) CAGTGGATAGACAGATGGGGG 25
MHCG2a CAGTGGATAGACCGATGGGGG 26
(21mer)
MHCG2b CAGTGGATGAGCTGATGGGGG 27
(21mer)
M HCG3 (21mer) CAAGGGATAGACAGATGGGGC 28
[00183] Five pl
aliquots from each PCR reaction were run on a 1%
agarose/TBE (pH 8.8) gel to determine which had produced a PCR product of
the correct size (ca. 450 bp). Those putative positive PCR products so
identified were directly cloned into the pCR2.1 vector provided by the TA
Cloning kit and transformed into TOP10 competent cells as described in the
manufacturer's protocol. Colonies containing the plasmid with a correctly
sized insert were identified by PCR-screening the colonies using the 1212 and
1233 oligonucleotide primers (Table 3) according to the method of GOssow
and Clackson (Nucleic Acids Res. 17:4000). Those putative positive clones so
identified were double-stranded plasmid DNA sequenced using the ABI
PRISM 310 Genetic Analyzer and the ABI PRISM BigDye TM terminator. Three
positive clones each of the VH and VK genes from the B2C4 hybridoma cell line

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
clone were sequenced, as were four positive clones of the VK gene and six of
the VH gene from the B2D6 hybridoma cell line clone.
Table 3 ¨ Primers for PCR screening and sequencing transformed colonies
Name Sequence (5'->31 SEQ ID
NO:
1212 (17m er) GTTTTCCCAGTCACGAC 29
1233 (21m er) AG C G GATAATTTCACACAG GA 30
[00184] The results of the 12 PCR reactions performed for each
hybridoma clone (B2C4 and BCD6) to amplify the murine 11-1F4 antibody
heavy chain variable region gene are presented in Table 4(a).
[00185] The degenerate leader sequence primer MHV7, in combination
with a mix of the MHCGI-3 constant region primers (Table 1), yielded a PCR
product of about 600 bp from template cDNAs derived from both the B2C4 and
B2D6 hybridoma cell lines. Since this band was larger than the expected size
for an average VH gene (450 bp), it was not investigated further. Conversely,
the degenerate leader sequence primer MHV6, in combination with a mix of
the MHCGI-3 constant region primers (Table 1), yielded a PCR product of the
expected size (450 bp) for a VH gene from template cDNA derived from both
the B2C4 and B2D6 hybridoma cell lines.
[00186] Table 4 shows the results of the PCR amplifications performed to
clone the murine 11-1F4 monoclonal antibody variable region heavy (a) and
light (b) chain genes from the 5P2/0 hybridoma cell lines B2C4 and B2D6.
Column three contains a record of the actual PCR results. Where a band was
observed for a particular combination of primers its size in base pairs (bp)
was
recorded in the appropriate space.
51

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
Table 4- Results of PCR amplification
ka.)
.... ,
C Re 'on Primer I Leader Primer ' Approxhnate Band Size (p)
B2C4 ' B2D6
, - -
. .
- ,
MHCGI-3 (mix) ME-1V1
WW2
_. .
it t MITV3 _ .
it MgV4--
it MHV5
.. _
- .._
sµ MEIV6 i 450 450
it ___________ MffV7 600 600
44. __________ tviaqr
., MEIV9
it MHV10 1
_ . . õ ___________________________________________________
-4H¨fir¨ 's
________ 44 mErvi2 _______
_ __________________________________________________________________________
,
'
(b)
CI, ftion Primer .r Leader Primer Approximate Band Size (bp)
1
132C4 B2D6
i
_ , ,
MKC MKV1 450 450
, ._. , .._
ti MKV2 <450' <450 .. 44, MKV3
_ .........................................................................
MKV4
_
¨ __________________________________________________________________________
,
it
=
¨ MKV5
44 NtKV6 200 1 .
it MXV7 ,
44
MKV8 t
....*-.
tt MKV9
44
MKV1 0 ... __
¨ .
.......¨*.....4........ ____________________________________________________
44 MK.V1 J.
....,....,...,, . ¨ . ¨ .=

' __________________________________________________________________________
,
52

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[00187] Sequence analysis of three clones from the B2C4 derived PCR
product and five clones from the B2D6 derived PCR product revealed a single
heavy chain variable region sequence (Figure 2).
[00188] The cloning strategy used (amplification of the entire variable
region gene by using primers which flank this region, i.e., leader sequence
and
constant region sequence specific primers) allowed the complete FR1
sequence to be identified. All eight clones sequenced had identical sequence
in this region (Figure 2).
[00189] The results of the 11 PCR reactions performed for each
hybridoma clone (B2C4 and BCD6) to amplify the murine 11-1F4 antibody
kappa light chain variable region gene are presented in Table 4(b).
[00190] The degenerate leader sequence primer MKV6 in combination
with the MKC constant region primer (Table 2), produced a PCR product of
about 200 bp from template cDNA derived from the B2C4 hybridoma cell line
only. Since this band was much smaller than the expected size for a VK gene
(450 bp), it was not investigated further.
[00191] The degenerate leader sequence primer MKV2, in combination
with the MKC constant region primer (Table 2), produced a PCR product which
was smaller than the expected 450 bp band (when viewed on an agarose gel)
from template cDNA derived from both the B2C4 and B2D6 hybridoma cell
lines. In addition, previous VK cloning had found that the MKV2 primer
amplified a well known kappa light chain pseudogene. Therefore, sequence
analysis of one clone of each PCR product was performed in order to confirm
that this product was a pseudogene and not the murine 11-1F4 antibody VK
gene. This sequence analysis revealed that this PCR clone was indeed the
pseudogene.
[00192] Finally, the degenerate leader sequence primer MKV1, in
combination with the MKC constant region primer (Table 1), produced a PCR
53

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
product of about the expected size (450 bp) for a VK gene, from template
cDNA derived from both the B2C4 and B2D6 hybridoma cell lines.
[00193] Sequence analysis of three clones of the B2C4 derived PCR
product and four clones of the B2D6 derived PCR product revealed a single
kappa light chain variable region sequence which could not be identified as a
pseudogene.
[00194] Thus, the 11-1F4 antibody heavy chain variable region gene was
cloned (using constant region specific and leader sequence specific primers)
from the hybridoma mRNA and sequenced.
[00195] When translated, the sequence gave a TVSS peptide sequence.
Analysis of 122 rearranged human VH genes, recorded in the Kabat database
(Kabat et al. - Sequences of Proteins of Immunological Interest), revealed
that
84% of these sequences had a TVSS peptide sequence. It was therefore
concluded that the VH gene isolated was the correct 11-1F4 antibody gene
sequence.
[00196] The murine 11-1F4 antibody variable region kappa light chain
gene was also successfully cloned and sequenced, as was a non-functional VK
pseudogene gene. This pseudogene was first identified by Carroll et al
(Molecular Immunology (1988) 25:991). The sequence arises from an aberrent
mRNA transcript which is present in all standard fusion partners derived from
the original MOPC-21 tumor (including 5P2/0). As a result of the aberrant
mRNA, the invariant cysteine at position 23 is replaced by a tyrosine residue,

and the VJ joint is out of frame, resulting in a stop codon at position 105.
[00197] It is common for lymphoid or hybridoma cells to synthesize more
than one rearranged light immunoglobulin mRNA. These mRNAs are usually
non productive due to the presence of termination codons or frame shifts not
usually seen in functional VK genes. These pseudo messengers often present
major problems when cloning immunoglobulin genes from hybridomas
because they are very good substrates for V region PCR, despite the fact that
they do not encode functional polypeptides.
54

CA 03071817 2020-01-31
WO 2019/027721 PCT/US2018/043374
[00198] The 11-1F4 antibody VK gene sequence was identified after
detailed sequence analysis of seven separate PCR clones, isolated from two
different PCR products to yield SEQ. ID NO: 36. Since all sequences were
identical, it was accepted as the correct 11-1F4 antibody kappa light chain
variable region sequence.
[00199] The cloned VH and VK region genes were used to make the
chimeric mouse-human 11-1F4 monoclonal antibody, which was then be
analyzed to confirm specific binding to AL fibrils.
EXAMPLE 2
Construction of chimeric mouse-human 11-1F4 (c11-1F4) antibody
[00200] In order to allow transient expression of the 11-1F4 VH and VK
variable region genes described above in mammalian cells as part of a
chimeric mouse -human antibody, it was necessary to modify the 5'- and 3'-
ends using specifically designed PCR primers (Table 5). The oligonucleotide
primers F39836 and F39837 were used to PCR modify the 11-1F4 VK gene,
while primers F39835 and F58933 were used to PCR modify the 11-1F4 VH
gene. The back (BAK) primers F39836 and F39835 introduced a HindlIl
restriction site, a Kozak translation initiation site, and an immunoglobulin
leader sequence to the 5' ends of the VK and VH genes respectively. The
forward (FOR) oligonucleotide primer F39837 introduced a splice donor site
and a BamHI restriction site to the 3' end of the VK gene while the
forward (FOR) oligonucleotide primer F58933 appended the first 22 base
pairs of the gamma-1CHi gene including an Apal restriction site to the 3' end
of the VH gene.
Table 5¨ Oligonucleotide primers used to PCR modify the 11-1F4 heavy
and kappa light chain variable region genes
Name Sequence 5' -> 3'
SEQ ID
NO:
F39835 AAGCTTGCCGCCAC CATGGCTGTCCTGGGGCTGCTC ITC 31
VH BAK TGC

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
F58933 CCGATGGGCCCTTGGTGGAGGCTGAGGAGACGGTGACT 32
VH FOR GAGGTTCC
F39836 AAGCTTGCCGCCACCATGAAGTTGCCTGTTAGGCTGTTG 33
VK BAK GTGC
F39837 GGATCCACTCACGTTTGATTTCCAGCTTGGTCCCCCCTCC 34
VK FOR GA
[00201] The Kozak consensus sequence is crucial to the efficient
translation of a variable region sequence (Kozak ¨ J Mol Bio 196:947). It
defines the correct AUG codon from which a ribosome commences translation,
and the single most critical base is the adenine (or less preferably, a
guanine)
at position -3, upstream of the AUG start.
[00202] The immunoglobulin leader sequence ensures that the
expressed antibody is secreted into the medium and therefore is easily
harvested and purified. The leader sequences used in this instance were the
murine 11-1F4 VK and VH leader sequences cloned from the hybridoma cDNA
during the VH and VK cloning process.
[00203] The splice donor sequence is important for the correct in-frame
attachment of the light chain variable region to its appropriate constant
region,
thus splicing out the 130 bp VK:CK intron. The heavy chain variable region was

attached directly to its appropriate constant region gene via the Apal site,
thus
eliminating the need for a splice donor site.
[00204] The sub-cloning restriction sites HindlIl and BamHI, and HindlIl
and Apal, respectively, bracket the modified VK and VH variable region genes,
while the use of different unique restriction sites ensured directional sub-
cloning into the appropriate mammalian expression vector.
[00205] The 11-1F4 light chain variable region gene was first carefully
analyzed to identify any unwanted splice donor sites, splice acceptor sites,
and
Kozak sequences (see Table 6). Both the heavy and light chain variable
region genes were analyzed for the presence of any extra sub-cloning
56

CA 03071817 2020-01-31
WO 2019/027721 PCT/US2018/043374
restriction sites which would later interfere with the subcloning and/or
expression of functional whole antibody. None were found.
Table 6 ¨ Sequences important for the efficient expression of
immunoglobulin genes in mammalian cells
Name Consensus DNA Sequences
Kozak translation initiation site CCGCCRCCAUGG
Kappa light chain splice donor site AC::GTRAGT
Heavy chain splice donor site AG::GTRAGT
Immunoglobulin splice acceptor site YYYYYYYYYYN CAG : :G
Bases shown in bold are considered to be invariant within each consensus
sequence.
[00206] Separate
PCR reactions were prepared as follows, one for each
variable region gene. The plasmids 11-1F4 VH.pCR2.1 and 11-
1F4
VK.pCR2.1 described above were used as templates. A 100 pl reaction
mixture was prepared in each PCR tube, each mixture containing up to 41p1 of
sterile water, 10 pI of 10 x PCR buffer I, 8 pI of the 10 mM stock solution of

dNTPs, 1p1 of 10 mM of 5' forward primer, 1p1 of the 10 mM 3' Reverse primer,
and 1p1 of a 1/10 dilution of template DNA. Finally, 0.5 pl of AmpliTaq DNA
polymerase (2.5 units) was added before overlaying the completed reaction
mixture with 50 pI of mineral oil. The reaction tubes were loaded into a DNA
thermal cycler and cycled (after an initial melt at 94 C for 1 min) at 94 C
for
30 sec, 68 C for 30 sec and 72 C for 50 sec over 25 cycles. The completion
of the last cycle was followed with a final extension step at 72 C for 7 min
before cooling to 4 C. A 10 pI aliquot from each PCR reaction tube was run
on a 1.2% (w/v) agarose/ 1 X TBE buffer gel containing 0.5 pg/ml ethidium
bromide to determine size and presence of a PCR- product. Positive PCR-
clones were about 420bp in size. Those putative positive PCR products so
identified were directly cloned into the pCR2.1 vector, provided by the Topo
TA
57

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
Cloning kit, and transformed into TOP10 competent cells as described in the
manufacturer's protocol. Colonies containing the plasmid with a correctly
sized
insert were identified by PCR-screening the colonies using the 1212 and 1233
oligonucleotide primers (Table 3) according to the method of GOssow and
Clackson. Those putative positive clones so identified were double-stranded
plasmid DNA sequenced using the ABI PRISM 310 Genetic Analyzer and
the ABI PRISM BigDye TM terminator. Two positive clones each of the Topo TA
cloned VH and VK genes were sequenced.
[00207] Clones containing the correctly modified 11-1F4 VH and 11-1F4
VK genes were identified and the modified V genes from these clones were
subcloned into their respective expression vectors to facilitate the
expression
of chimeric heavy and kappa light chains in mammalian cells. The modified
11-1F4 VK gene was subcloned into the expression vector pKN100
(Figure 4) as a HindIII-BamHI fragment; this vector contains a human kappa
constant region gene (allotype: Km (3 Ala153, Ser191)). The modified 11-1F4
VH gene was also subcloned as a HindIII-Apal fragment into the expression
vector pG1D200 (Figure 5); this vector contained a human y1 constant region
gene (allotype: G1m (-1 Glu377, Met38I, -2 Ala462, 3 Arg222, 5er229)). Both
the kappa and y1 constant region allotypes used are commonly found in the
caucasian population. The ligated expression constructs, 11-1F4VK.pKN100
and 11- 1F4VH.pG1D200, were then used to transform DH5a competent cells,
and positive clones were identified using the PCR screening method
discussed above with the original PCR modification primers (Table 4). The
expression vectors are readily available.
EXAMPLE 3
Construction of a single supervector for transient expression of chimeric 11-
1F4
in COS Cells.
[00208] A single supervector expressing both immunoglobulin chains of
the chimeric 11-1F4 antibody was constructed as follows. The 11-1F4 kappa
light chain expression cassette (which contained the HCMVi promoter, the 11-
58

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
1F4 kappa light chain variable region gene, and the kappa light chain constant

region gene) was restriction enzyme digested (EcoRI at positions 1 and
2490) out of the 11-1F4VK.pKN100 construct (Figure 4) and subsequently
ligated into the 11-1F4VHpG1D200 construct via the unique EcoRI (position
4297, Figure 5). This ligation resulted in the construction of a supervector
construct, pG1KD200-11-1F4, containing both the heavy and kappa light
chains of the 11-1F4 chimeric antibody.
EXAMPLE 4
Transient expression of the chimeric y1/K.11-IF4 whole antibody in COS cells
[00209] The chimeric 11-IF4 antibody was transiently expressed in COS
cells from the European Collection of Cell Cultures (ECACC) in two ways:
(i) By cotransfection of 10pg of each of the vector constructs 11-
1F4VK. pKN100 and 11-1F4VH.pG1D200. Co-transfections were carried
out in duplicate.
(ii) By transfection of 13pg of the single supervector construct
pG1KD200-11-1F4. Supervector transfections were carried out five times.
[00210] The following transfection method was used. The COS cell line
was grown in DMEM supplemented with 10% (v/v) FCS, 580 pg/ml L-
glutamine and 50 Units/ml penicillin/ 50 pg/ml streptomycin ("media") in a 150

cm2 flask until confluent. The cells were trypsinized, spun down in a bench
top
centrifuge (250 g for 5 min), then re-suspended in 6 ml of media before
dividing them equally between three 150 cm2 flasks, each containing 25 ml of
fresh, pre-warmed media. The cells were incubated overnight at 37 C in 5%
CO2 and then harvested the next day while they are still growing
exponentially.
Each flask contained approximately 1 x 107 cells. The cells were trypsinized
again, pelleted as before, and washed in 20 ml of PBS, following which they
were re-suspend in sufficient PBS to create a cell concentration of 1 x 107
cells/ml. 700 pl of these washed COS cells were pipetted into a Gene Pulser
cuvette, to which was then added 1 pl of both the heavy chain and kappa light
59

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
chain expression vector DNA (each at 10 pg) or 13 pg of the super-vector
construct. A 1900 Volt, 25 pFarad capacitance pulse was delivered to the
mixture using the Bio-Rad Gene Pulser apparatus. The pulsing was
repeated for each experimental transfection and a "no DNA" control (in which
the COS cells were electroporated in the absence of any DNA). A positive
control of a previously-expressed antibody was also carried out to test the
efficiency of the COS cells.
[00211] The COS cells were allowed to recover at room temperature for
min, then gently pipetted the into a 10 cm diameter tissue culture dish
containing 8 ml of pre- warmed DMEM supplemented with 10% (v/v) y -
globulin free FBS, 580 pg/ml L- glutamine and 50 Units/ml penicillin / 50
pg/ml streptomycin, and incubated in 5% CO2 at 37 C for 72 hours before
harvesting the COS cell supernatant for analysis. After incubation for 72
hours
the medium was collected, spun to remove cell debris and analyzed by ELISA
for chimeric antibody production and antigen binding of the c11-1F4 antibody.
EXAMPLE 5
Quantification of the chimeric y1/k 11-1F4 antibody via capture ELISA
[00212] Following expression, the whole IgG molecules present in the
COS cell supernatant were quantified using a capture ELISA assay. IgG
molecules were captured on a Nunc-Immuno MaxiSorbTM plate via an
immobilized goat anti-human IgG, Fey fragment - specific antibody, and
detected via an anti-human kappa light chain peroxidase conjugated antibody.
A standard curve was generated by capturing and detecting known
concentrations of a standard IgG antibody on the same plate in the same way
as follows. Each well of a 96-well immunoplate was coated with 100 pI
aliquots of 0.4 pg/ml goat anti-human IgG antibody diluted in PBS and
incubated overnight at 4 C. The excess coating solution was removed and
the plate was washed three times with 200 p1/well of washing buffer (1xPBS,
0.1% TWEEN). Into all wells except the wells in column 2, rows B to G, was
dispensed 100p1 of SEC buffer. A 1pg/m1 solution of the human IgG1/kappa

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
antibody in SEC buffer was prepared to serve as a standard and 200 p1/well
was pipetted into the wells in column 2, rows B and C. The medium from the
transfected cos cells was centrifuged (250g, 5 min), saving the supernatant.
An aliquot of 200 pl of the supernatant from the "no DNA" control (in
which COS cells were transfected in the absence of DNA) was pipetted into
the well in column 2, row D, and aliquots of 200 p1/well of experimental
supernatants were pipetted into the wells in column 2, rows E, F, and G. The
200 pI aliquots in the wells of column 2, rows B to G were mixed and then 100
pI was transferred from each well to the neighboring well in column 3. This
process was continued to column 11 with a series of 2-fold dilutions of the
standard, control, and experimental samples, following which all were
incubated at 37 C for 1 hour and all the wells were rinsed six times with 200

pI aliquots of washing buffer. The goat
anti-human kappa light chain
peroxidase conjugate was diluted 5000-fold in SEC buffer and 100 pI of the
diluted conjugate added to each well, followed by a repetition of the
incubation
and rinsing steps. To each well was added 150p1 of K-BLUE substrate,
followed by incubation in the dark at 25 C for 10min. The reaction was
stopped by adding 50 pl of RED STOP solution to each well and the optical
density was read at 655nm.
EXAMPLE 6
Binding analysis of the chimeric 11-1F4 antibody
[00213] The
chimeric 11-1F4 antibody was tested for binding to amyloid
fibrils using a direct binding ELISA assay. Synthetic fibrils were formed from

an immunoglobulin light chain protein and used to monitor the reactivity of
the
antibody in a solid-state ELISA-based assay using a "low- binding" polystyrene

plates (Costar,# 3474). Immediately prior to coating the plate, a mass of 250
pg of fibrils was diluted to 1 ml with coating buffer (0.1% bovine serum
albumin in phosphate buffered saline pH 7.5). The sample was then sonicated
for 20 sec using a Tekmar Sonic Disruptor sonicating probe, with the power
set to 40% of maximum, resulting in a solution of short fibrils composed of up
61

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
to 2-5 protofiliments each. This solution was then diluted to 5 ml, mixed well
by
vortex, and aliquoted into the wells of the plate. This process yielded 50 pl
of
fibril solution having a concentration of 50 pg/ ml in each well. The plate
was
then dried overnight by placing it uncovered in a 37 C incubator.
[00214] The ELISA
assay was then performed as follows within 48 hours
of preparing the plate. The wells were blocked by the addition of 100 pl of I%

BSA in PBS and incubated for 1 hour at room temperature on a shaker. The
plate was washed x3 in PBS, 0.05% Tween 20 (v/v). To each well of the plate
was added 50 pI of a solution of c11-1F4 (3pg/m1 antibody in 0.1% BSA/PBS)
and the plate incubated at room temperature for 1 hour on a shaker. The plate
was again washed x3 (as before) and detection of bound antibody was
accomplished using a biotinylated goat anti-mouse IgG antibody (Sigma# B-
8774, anti-heavy and light-chain).
Results
[00215] Sequence
analysis of the successfully modified VH and VK genes
revealed the correct sequence was present. Detailed DNA and amino acid
sequences of the modified 11-1F4 VK and VH genes are presented in Figures 3
& 4. The modified VK and VH genes were successfully cloned into the
mammalian expression vectors pG1D200 and pKN100 respectively, and the
resulting 11-1F4VK.pKN100 and 11-1F4VHpG1D200 constructs were used for
cotransfection of mammalian cells.
[00216] The 11-
1F4VK. pKN 100 and 11-1F4VHpG1D200 constructs were
also subsequently used to construct a single supervector (pG1KD200-11-1F4),
which expressed the chimeric 11- 1F4 antibody in mammalian cells. The
chimeric II-1F4 antibody expression levels, from both cotransfections and
supervector transfections of ECACC COS cells were assayed. The
expression levels observed from the pG1KD200-11-1F4 supervector
transfections (10326 ng/ml) were 3.7 fold higher than the levels observed from

the corresponding co-transfections of the 11-1F4VK.pKN100 and 11-
1 F4VHpG1D200 constructs (2820 ng/ml).
62

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[00217] Following expression and quantification, the chimeric 11-1F4
antibody was tested for binding to target antigen (amyloid fibrils kindly
supplied
by the NCI) by direct binding ELISA. The results of the binding ELISA are
presented in Figure 8. Supenatants from the two best individual pG1KD200-
11-1F4 supervector transfections were assayed in parallel with one
supernatant from the corresponding co-transfection.
[00218] The results indicated that the chimeric 11-1F4 antibody bound to
the amyloid fibrils with a higher affinity than its murine equivalent. This
result is surprising and unexpected because normally a chimeric antibody
would be expected to have a binding affinity comparable to the original murine

antibody. Without intending to be bound by the particular mechanism, the
inventors believe it is possible that the net effect of combining the 11-1F4
murine V regions with the human y1/k C regions used to create the chimeric
11-1F4 antibody produced an antibody of higher affinity.
[00219] Samples of CHO cells (identified as CAEL-101) that secrete the
chimeric 11-1F4 monoclonal antibody used herein were deposited with the
American Type Culture Collection (ATCC Acc. No: PTA-125146) on June 27,
2018, in compliance with the Budapest Treaty.
EXAMPLE 7
Mouse Amyloidoma Study with Murine 11-1F4
[00220] Amyloids were extracted from humans and characterized.
Briefly, 30 to 40 g of fresh-frozen (-80 C) or 10 g of lyophilized spleen or
liver
obtained postmortem from patients with AL amyloidosis were homogenized in
-300 ml of cold saline with a Virtis-Tempest apparatus (Virtis, Gardiner, NY).

The homogenates were centrifuged at 6 C for 30 minutes at 17,000 rpm and
residual saline-soluble material was removed by repeated homogenization and
washing until the resultant supernatant had an OD of <0.10 at A280. The pellet

was then repeatedly homogenized, washed with cold deionized water,
centrifuged, and the amyloid-containing supernatants lyophilized. The amount
of protein recovered represented approximately one-third to one-fifth the
63

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
weight of the starting material. The light chain composition and VL subgroup
of
the amyloid was determined by amino acid sequencing (Procise Protein
Sequencing System; Applied Biosystems, Foster City, CA) and ionizing mass
spectroscopy (PE SCIEX API 150 EX; Perkin Elmer, Norwalk, CT) of high-
performance liquid chromatography-separated peptides obtained by trypsin
digestion of reduced and pyridylethylated protein extracted from the water-
soluble material with 6 mol/L guanidine HCI. The presence of the proteoglycan
heparan sulfate was established using an Azure Aassay.
[00221] The composition of the amyloid extracts was established by
chemical, immunoblotting, amino acid sequence, and ionizing mass
spectroscopic analyses where the predominant protein species were found to
be K or A light chain-related molecules that, in most cases, consisted
primarily
of the variable region (VL) plus the first -50 residues of the constant region

(CL) and, in others, VL fragments or intact molecules. Additionally, these
extracts contained the expected amyloid-associated P- component, as well as
the proteoglycan heparin sulfate.
[00222] Lyophilized water-soluble amyloid extracts were suspended in 25
ml of sterile saline and homogenized with a PCU-2 Polytron apparatus
(Brinkman, Luzerne, Switzerland). The fibrils were sedimented by
centrifugation at 6 C for 30 minutes at 17,000 rpm; the resultant pellet was
resuspended in 1 ml of sterile saline and rehomogenized. This solution was
injected subcutaneously between the scapulae of BALB/c, CD-18 null, and
C.B-17 SCID mice using an 18-gauge needle attached to a 6-ml syringe. The
size of the resultant amyloidoma was measured by daily palpation and
confirmed at necroscopy. High-resolution X-ray-computed tomography images
were acquired using a microCat apparatus (Oak Ridge National Laboratory,
Oak Ridge, TN).
[00223] The injected material formed a readily visible, palpable mass on
the backs of animals, the size of which depended on the amount of material
injected (e.g., 0.2 to 2.5 cm in maximum diameter). The amyloidoma remained
64

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
localized and unchanged for -10 to 24 days, as evidenced by high-resolution
X-ray-computed tomography; after that point, the amyloidomas began to
regress and eventually disappeared throughout an -4-day period. This
response occurred regardless of the K or A nature or the VL subgroup of the
amyloid extract; however, in studies involving five different K and seven A
amyloidomas, ALA extracts typically resolved more slowly than did ALk (ALA,
18 +/- 6 days versus ALK, 13 +/- 3 days). Sufficient material was available to

repeat experiments at least four times in eight of the 12 cases where it was
found that this effect was reproducible in healthy, young animals regardless
of
the tissue source of the amyloid. However, dissolution of the induced
amyloidomas was consistently delayed beyond 3 months in aged (>18
months) and immunodeficient mice.
[00224] Histological studies to determine the fate of the regressing
amyloidomas demonstrated that the amyloid was not redistributed to other
mouse tissues, as evidenced by Congo red staining. Additionally, the
amyloidomas were infiltrated by naphthol AS-D chloroacetate-positive, a-
naphthyl acetate-negative, polymorphonuclear cells, i.e., neutrophils. In
contrast, this cellular response did not occur in CD-18 null mice where
resolution of human AL amyloidomas required a considerably longer time
period (i.e., ;3 months). Further, amyloidolysis was delayed in animals
rendered profoundly neutropenic by co-administration of 250 mg of the anti-
neutrophil mAb Gr-1 given at the time of amyloidoma induction and again on
day 3.
[00225] Amyloid removal also was dependent on a humoral murine
response to the human light-chain-containing material. Approximately 10 to 20
days after amyloidoma induction, we showed in immunoblotting experiments
that mouse sera contained antibodies that recognized, not only the light chain

constituent of the amyloid protein injected, but also that of heterologous ALK
or
ALA extracts. In contrast, there was no reactivity with the homologous amyloid

precursor protein, i.e., Bence Jones protein or any other monoclonal light
chain tested. When the same amyloid preparation was re-administered to

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
these immunized animals, its rate of disappearance increased approximately
twofold.
[00226] To test
the therapeutic efficacy of murine 11-1F4, a series of
experiments were initiated in which 100-pg doses of the antibody were given
to pairs of mice bearing human AL amyloidomas. In the case of ALk, studies
involving two different extracts revealed that even a single injection of the
antibody resulted in rapid and complete disappearance of the amyloid tumor,
as compared to untreated animals (Table 7; Figure 9). The mass of an ALkA
amyloidoma was reduced >90% within 4 days after antibody injection, as
compared to control animals. However, to achieve a similar response in
certain ALA-type amyloidomas, multiple doses of the reagent were required.
These were given as a series of 100-pg injections beginning at the time when
the amyloidoma was induced (day 0) and then again on days 2, 4, and 6
(Figure 9B). As summarized in Table 7, in experiments in which five different
human ALA amyloidomas were tested in the mouse model, it was found that
treatment with 11-1F4 decreased by as much as fourfold the time in which the
amyloid tumors were eliminated. Notably, although single or repeated doses of
two other anti-light chain mAbs that recognized AL fibrils (e.g., 31-8C7)
expedited amyloidolysis, the 11-1F4 reagent was unique in that it accelerated
removal of both ALk and ALA amyloid, albeit at different rates. In contrast,
three other anti-light chain mAbs that were tested lacked such activity.
Table 7 ¨ Treatment of Mice with Human Amyloidomas with Murine 11-1F4
SneKaAi.z.Ae
Treated Trealed
AnlyIoluortia
('4 su)gritip) 31-807' Untreeted 11-1t4 31-8C7
lintreated
ALK( 1) KG 12 14 Isfr NT NT
ALK(1) GRA 4 18 NT NT
ALA(6) JON B 11 14 NT NT NT
AL) SHE 9 19 21 9 20 24
.ALAC1) FIE 1? 24 24 9 19 26
ALA(2) BUE 24 2i 6 NT 25
ALA(3) BAL 28 NT 28 7 NT 28
'100 day i3; 1100 Kt) days 0.2. 4, and Ei: frnAt: designafion: '5tim _=
(dt.P13); 9t4T. TrOt SOSted.
[00227] It was
also determined that 11-1F4 recognized other forms of
amyloid, as evidenced in immunohistochemical analyses of AA-, ATTR-,
66

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
ALyS-, AApoAl -, and Ab-containing tissues. In each case, similar patterns of
reactivity were obtained with 11-1F4 and antibodies specific for these five
different types of amyloid proteins.
EXAMPLE 8
Phase la/b Study of the chimeric 11-1F4 antibody
[00228] GMP-grade amyloid fibril-reactive Chimeric IgG1 mAb 11-1F4
was produced by NCI's Biological Resource Branch for a phase 1 a/b trial
treating patients with refractory AL amyloidosis. The CHO cell line producing
the chimeric IgG1 mAb 11-1F4 is identified as CAEL-101.
[00229] Relapsed or refractory AL Amyloidosis patients who received
prior anti-plasma cell treatment were enrolled. Patients received chimeric
IgG1
mAb 11-1F4 as a single intravenous infusion (phase 1a) or a series of weekly
infusions for 4 weeks (phase 1b). A dose-escalation "up and down" design was
used for both phase la and lb where successive doses of 0.5, 5, 10, 50, 100,
250 and 500 mg/m2 were administered.
[00230] The primary objective of the study was to establish the maximum
tolerated dose of chimeric 11-1F4, and secondary objectives included: (1)
demonstrating a reduction in amyloid burden as evidenced by a decrease in
affected organomegaly and/or improved organ function; (2) determining the
pharmacokinetics of 11-1F4 when given as a single IV infusion (phase 1a) or
as a series of weekly IV infusions (phase lb); and (3) determining the
difference between 250 mg/m2 and 500 mg/m2 doses.
[00231] Key inclusion criteria included being 21 years of age or older,
the
patient had previously received systemic therapy, the patient did not require
plasma cell targeted therapy, and the patient had an Eastern Cooperative
Oncology Group (ECOG) performance status of less than or equal to 3.
[00232] Key exclusion criteria included intraventricular septum of greater

than 2.5 mm, creatine clearance less than 30 cc/min, alkaline phosphatase
67

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
more than 3 times the institutional upper limit of normal, and bilirubin
higher
than 3.0 mg/dL.
[00233] For the
Phase la study, dose escalation followed an "up and
down design." Once
tolerated, successive patients each received
progressively higher doses of chimeric 11-1F4 mAb, with two patients enrolled
at a dose of 500 mg/m2. Even the patients receiving the 500 mg/m2 dose did
not report any dose limiting toxicities. Patients were evaluated at week 0,
dosed with chimeric 11-1F4 at week 1, and then reevaluated at weeks 2, 3, 4,
and 8, as shown in Figure 9A.
[00234] For the
Phase lb study, infusions were given once a week for
four weeks starting at 0.5 mg/m2, Once tolerated, successive patients each
received progressively higher doses of chimeric 11-1F4 mAb, with six patients
enrolled at a dose of 500 mg/m2. The dosing scheme is shown in Figure 9B
Results
[00235] Twenty
seven patients were treated with chimeric 11-1F4A
antibody. Twenty six patients were evaluable for response. Eight patients
completed phase la and nineteen patients completed treatment in phase lb.
Median age for Phase la and lb was 68. All patients tolerated the given dose
of mAb chimeric 11-1F4 and up to the highest dose level of 500 mg/m2 for
both phase 1 a and 1 b. There were no drug-related grade 4 or 5 adverse
events (AEs) or dose-limiting toxicities. Two patients developed a grade 2
rash
3-4 days after infusion. One patient developed the skin rash in in phase 1 a
(dose level 4) and when he was retreated in phase 1 b. A skin biopsy with
immunohistochemical staining showed chimeric 11-1F4 binding to amyloid
fibrils with an concomitant neutrophilic infiltrate. The same patient and
another
patient developed a similar rash in phase lb which further provides clinical
and
correlative data that chimeric 11-1F4 directly binds to light-chain amyloid
fibrils. Overall, 63% (5 of 8) of evaluable patients demonstrated organ
response after one infusion of mAb cl 1-1F4 in phase 1a. The median time to
response in phase la was 4.5 weeks after completing therapy. In phase lb,
68

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
61% (11 of 18) of evaluable patients showed significant organ responses with
a median time to response of 1 week after commencing therapy with the
tendency of faster response in higher dosages
[00236] The patient characteristics of a subset of the evaluable patients
are shown in Table 8 below.
Table 8 - Phase la/b Patient Characteristics
Characteristic Median
Age (N=21 67 yrs
patients) (Range: 34 ¨
77)
Gender Male N=15 (68%)
Female N=6 (32%)
Light Chain A N=13 (52%)
type K N=8 (48%)
Revised Mayo II (Range: Ito
Stage IV)
Organ 2 (Range: 1 ¨ Heart N=11 (52%)
Involvement 4) Kidney N=11 (52%)
(No.) Skin/Soft tissue N=10 (48%)
GI N=8 (38%)
Nervous system N=4 (19%)
Liver N=3 (14%)
Lung N=2 (10%)
Musculoskeletal N=1 (5%)
Best CR N=3 (14%)
Hematologic VGPR N=15 (71%)
Response to PR N=2 (10%)
Therapy NR N=1 (5%)
Previous 2 (Range: 1 ¨
Regimen (No.) 6)
Baseline NT- 2359
proBNP (Range: 894 ¨
(ng/L)a 13,131)
Baseline 24 hr 4998
Urine Protein (Range: 1078 ¨
(mg/24hr)b 10,170)
Time Since 6 (Range 1 ¨
last Exposure 51)
to
Chemotherapy
69

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
(MOs)
a Baseline NT-proBNP in patients with cardiac involvement who were evaluable
for response (Baseline NT-proBNP> 650pg/mL)
b Baseline 24 hour urine protein in patients with renal involvement who were
evaluable for response (Baseline 24 hour urine protein > 500mg/24 h)
[00237] At the close of the Phase la/b studies, 18 patients had evaluable
responses (N=1 had no measurable disease, N=2 did not complete treatment).
Twelve of the 18 (67%) showed an improved organ response. Specifically, in the

Phase la, 63% of patients (5 of 8) with measurable disease burden
demonstrated organ response after one infusion of mAb 11-1F4 (2 renal, 2
cardiac, and 1 GI). In the Phase lb study, 70% of patients (7 of 10) with
measurable disease burden showed organ response: 3 of 4 patients who were
evaluated for response with cardiac involvement showed cardiac response; 4 of
4 patients who were evaluated for response with renal involvement showed renal

response; 1 patient with GI response was evaluated; and 1 patient with soft
tissue response showed an improvement of arthritis from 3 4 1.
Cardiac Response
[00238] Eight patients were evaluated for cardiac response. Among the
metrics that were evaluated were NT-proBNP and NYHA class criteria. The
baseline level for all of these patients was 650 pg/ml. Five of the patients
(63%)
showed a significantly improved response (i.e., a 30(:)/0 decrease in NT-
proBNP
and/or shifting from NYHA class III to class I), 2 patients remained stable,
and
only one showed any sign of disease progression. The cardiac results for the
groups of patients are shown in Figure 11. The decrease in NT-proBNP for one
exemplary patient is shown in Figure 12.
Renal Response
[00239] Eight patients were evaluated for renal response, with proteinuria

being the primary metric for determining responsiveness. Six patients (75%)
showed a significantly improved response (i.e., a 30(:)/0 decrease in
proteinuria
or a decrease to <0.5 g/24 hours in the absence of renal progression), and two

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
patients remained stable. No patients showed signs of renal disease
progression (>25% worsening in eGFR). The renal results for the groups of
patients are shown in Figure 13. The decrease in proteinuria for one
exemplary patient is shown in Figure 14.
Study Overview of Results
[00240]
Treatment with chimeric 11-1F4 was well tolerated and safe. There
were no drug related grade 4 or 5 adverse events (AEs) or dose limiting
toxicity
up to an MTD of 500mg/m2. Moreover, chimeric 11-1F4 is clinically efficacious.

Most patients saw an early and sustained organ response even as a single
infusion or as a weekly infusion for 4 weeks. Improved responses were observed

across tissues/organs, including cardiac, renal, GI, skin, and soft tissue
responses. Indeed, chimeric 11-1F4 safely promotes amyloid resolution in 67%
of the patients and leads to improvement in organ function after just a single

dose, even in patient with ALA deposits. Patient response to chimeric 11-1F4
was rapid and sustained. Indeed, with median response time of 4.5 weeks in the

Phase la trial and just one week in the Phase lb trial, chimeric 11-1F4
provides
a positive response faster than any other known therapeutic targeting amyloid
fibrils. The rapid destruction of amyloid fibrils by chimeric 11-1F4 can
improve
organ function and, by extension, significantly improve mortality in patients
with
this uniformly fatal disease.
EXAMPLE 9
Cardiac Response to Chimeric Fibril-Reactive Monoclonal Antibody 11-1F4 in
Patients with AL Amyloidosis with Global Longitudinal Strain: Results from the

Phase 1 b Trial
[00241] An open-
label phase lb clinical trial of the chimeric 11-1F4 mAb
was completed with promising results as shown below. This study was
undertaken to assess the response of myocardial function to mAb administration

using global longitudinal strain (GLS).
71

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
[00242]
Nineteen patients with relapse or refractory AL Amyloidosis were
enrolled into the trial (age SD, 63 12; 68% male). Fifty three percent had
light
chain kappa amyloid and 52% had cardiac involvement as defined by NTpro-
BNP level of > 650 pg/ml. NTpro-BNP screening and baseline levels of the
nineteen patients are shown in Table 9 below. These cardiac patients included
2
patients who were not in the cardiac evaluable primary clinical analysis, due
to
differences in Screening vs Baseline NT-proBNP values.
Table 9 - NTpro-BNP Screening and Baseline Values of Echocardiographic
Analysis
Visit Statistics All Phase 1B Cardiac Cardiac
patients involvement Evaluable
Screening n 19 10 B
Mean (SD) 992.81 (1067.07) 1681.4 1846.9
(1067.12) (1127.46)
11111646011001=00212Millik.,.. 11866 15926
Min, Max 39.7, 3964.0 662.2, 3964.0 850.1, 3964.0
Baseline n 19 10 B
(Week 1)
1111111111111111111111111111111111111111111111111111111111111111111111111111111
1111111111111111111
Mean (SD) 897.95(1067.81) 1560.9 1796.9
(1114.99) (1131.27)
Medan 5S970 9B640 1261 0
Min, Max 44.1, 3810.0 589.7, 3810.0 815.5, 3810.0
[00243] The mAb
was administered weekly for 4 weeks with sequential
doses of 0.5, 5, 10, 50, 100, 250 and 500 mg/m2 in a dose-escalation design.
Clinical echocardiographic (ECHO) examinations at baseline and 12 weeks post
therapy were compared. Several echocardiographic variables including left
ventricular ejection fraction (LVEF) (calculated using Simpson's biplane
method)
and global longitudinal strain (GLS) were obtained. GLS was measured using
speckle-tracking (TomTec-Arena 1.2, Germany) and calculated as an average of
4-, 2-, and 3- chamber based measurements. Paired student's t-test was used to

compare echocardiographic variables at baseline and 12 weeks after therapy
with mAb. The analysis of the echocardiogram parameters are demonstrated in
Table 10 below.
72

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
Table 10 - The analysis of the echocardiographic parameters
Screening. Week 12 P-vaIne
Number Patients 1.0 10
Ividd....em (mean (sd)) 4.38 (0.93) 4.32 (0.91) 0319
ivs_cm (mean. (sd)) 1.29 (0.22) 1.21 (0.18)
pwt_cm (mean (sd)) 1.12 0.27) 1.14 0.25) 0.2.17
Ived....tnass...g (mean (sd)) 187.86 (4337) 179.70 (4987)
0..197
ef_percent (mean (sd)) 51.95 0.92) 52.28 (11.59)
0..856
mcf (mean (.sd)) 0.37 ((U1) 0.38 (0.13) 0.626
mv_e_m_s(m.ean (sd)) 8.53 (24.18) 0.94 (0.37) 0.347
Inv a m s.ancan (sd)) 13.45 (34.05) 0.72 (0.23) NA
gls_percent (mean (sd)) -15.68 (4214) -17.37 (3.53) 0..004
[00244] While
there was neither significant change between LVEF (56.2
8.6% vs. 56.2 9.5%, p = 0.985) nor GLS (-19.04 -5.11% vs. -19.73 -4.1%,
p
= 0.119) from baseline to 12 week examinations for the overall cohort,
patients
with cardiac involvement demonstrated an improvement in GLS (-15.58 -4.14%
pre and -17.37 -3.53% post, p = 0.004 as visualized in Figure 15. An
exemplary echocardiogram of a patient with cardiac involvement before
treatment and at week 12 post treatment with chimeric 11-1F4 mAb is shown in
Figure 17. The patient shown in Figure 17 had a baseline level of NT-proBNP of

2549 pg/mL and a GLS value of -9.58 before treatment. After 12 weeks of
chimeric 11-1F4 mAb treatment, the patient exhibited reduction in GLS to -
13.39,
73

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
and a reduction in NTproBNP to 1485 pg/mL. Subgroup analysis showed an
improvement in GLS in patients with lambda amyloid cardiac involvement (- 14.3

- 4.38% pre and - 16.17 -3.74% post, p = 0.02) and a trend in improvement
with kappa amyloid cardiac involvement (-16.60 - 4.10% pre and -18.16 -
3.48% post, p = 0.07). Furthermore, the Cardiac Evaluable Population as
defined
in the clinical analysis of the study (using the Baseline NT-proBNP values
rather
than the Screening Values) also resulted in a statistically significant
decrease in
GLS% (p-value 0.0163), with a numerically similar decrease (-1.71) as the
analysis provided by the ECHO group (-1.69). Table 11 below shows the analysis

of decrease in the GLS in cardiac patients versus cardiac evaluable patients
and
non-cardiac patients.
Table 11 - Comparison of Cardiac Patients per Screening vs Baseline
Evaluable Definitions and Non Cardiac Patients.
Number Patients Baseline P-vaine-
rOLS 10 (Cardiac Patients per -15358 (4.14)
47,37 (3,53) 0,004
Mean (Std Seremin,g NT-prof3NP)
=
Dev)
8 (Cardiac Evaittabie per -14,95 (.432) -16.66 (3,55)
0.0163
Baseline NT-proBNP)
9 (Non-Cardiac Patient& per -22.77 (3.12) -22.36 (.3.02)
0.4829
Screening NT-proBNP)
[00245] In conclusion, this trial shows a significant improvement in GLS
after exposure to an anti-fibril specific mAb in subjects with AL amyloid
cardiac
involvement. As shown in Figure 15, 9 out of 10 patients with cardiac
involvement improved on GLS%. The probability that 9 or more patients
improving, under the null hypothesis that there is no drug effect, is -0.0107,

suggesting that observing 9 of 10 patients improving is a highly unlikely
outcome
unless the drug truly is effective. This preliminary data will aid in
designing a
larger clinical trial. Additionally, larger trials leveraging GLS to evaluate
myocardial function are warranted.
EXAMPLE 10
74

CA 03071817 2020-01-31
WO 2019/027721
PCT/US2018/043374
Organ Response to Chimeric Fibril-Reactive Monoclonal Antibody 11-1F4 in
Patients Who Are Not Controlled Hematologically.
[00246] A patient that had received 6 chemotherapy treatments and had
achieved a hematologic partial response on those treatments with no organ
response was administered the chimeric amyloid fibril-reactive monoclonal
antibody (mAb) 11-1F4. For three consecutive periods post-dose, there was
consistent reduction in NT-proBNP after receiving 11-1F4, and the patient
achieved an organ response as described in Figure 16. But when taken off of
antibody, free light chains increased and the patient condition worsened.
Organ
progression was then seen after completion of the trial. The response pattern
of
this patient led the investigators to conclude that organ response was due to
the
chimeric 11-1F4 antibody treatment and independent of chemotherapy induced
hematologic response.
[00247] In the description and claims of this specification the word
"comprise" and variations of that word, such as "comprises" and "comprising"
are not intended to exclude other features, additives, components, integers or

steps but rather, unless otherwise stated explicitly, the scope of these words

should be construed broadly such that they have an inclusive meaning rather
than an exclusive one.
[00248] Although the compositions and methods of the invention have
been described in the present disclosure by way of illustrative examples, it
is
to be understood that the invention is not limited thereto and that variations

can be made as known by those skilled in the art without departing from the
teachings of the invention defined by the appended claims.

Representative Drawing

Sorry, the representative drawing for patent document number 3071817 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2018-07-24
(87) PCT Publication Date 2019-02-07
(85) National Entry 2020-01-31
Examination Requested 2020-01-31

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $100.00 was received on 2024-06-24


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if standard fee 2025-07-24 $277.00 if received in 2024
$289.19 if received in 2025
Next Payment if small entity fee 2025-07-24 $100.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee 2020-01-31 $200.00 2020-01-31
Request for Examination 2023-07-24 $400.00 2020-01-31
Registration of a document - section 124 $100.00 2020-05-04
Maintenance Fee - Application - New Act 2 2020-07-24 $50.00 2020-07-13
Maintenance Fee - Application - New Act 3 2021-07-26 $50.00 2021-06-23
Maintenance Fee - Application - New Act 4 2022-07-25 $50.00 2022-07-11
Maintenance Fee - Application - New Act 5 2023-07-24 $100.00 2023-07-10
Maintenance Fee - Application - New Act 6 2024-07-24 $100.00 2024-06-24
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
THE TRUSTEES OF COLUMBIA UNIVERSITY IN THE CITY OF NEW YORK
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2020-01-31 1 58
Claims 2020-01-31 12 429
Drawings 2020-01-31 14 608
Description 2020-01-31 75 3,552
Declaration 2020-01-31 1 10
National Entry Request 2020-01-31 8 174
Voluntary Amendment 2020-01-31 6 218
Non-compliance - Incomplete App 2020-02-12 2 205
Cover Page 2020-03-25 1 32
Claims 2020-02-01 4 152
Completion Fee - PCT 2020-05-04 8 274
Maintenance Fee Payment 2020-07-13 1 33
Examiner Requisition 2021-02-24 4 234
Amendment / Sequence Listing - Amendment / Sequence Listing - New Application 2021-06-24 40 2,730
Claims 2021-06-24 4 176
Description 2021-06-24 75 3,678
Examiner Requisition 2022-02-16 3 161
Claims 2022-06-10 4 173
Amendment 2022-06-10 11 396
Examiner Requisition 2023-01-09 3 179
Amendment 2023-05-05 11 362
Claims 2023-05-05 4 219
Office Letter 2024-03-28 2 189

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :